CN116019819A - Active ginsenoside composition and preparation method and application thereof - Google Patents

Active ginsenoside composition and preparation method and application thereof Download PDF

Info

Publication number
CN116019819A
CN116019819A CN202211636359.9A CN202211636359A CN116019819A CN 116019819 A CN116019819 A CN 116019819A CN 202211636359 A CN202211636359 A CN 202211636359A CN 116019819 A CN116019819 A CN 116019819A
Authority
CN
China
Prior art keywords
active
composition
ginseng
ginsenoside
total
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202211636359.9A
Other languages
Chinese (zh)
Inventor
连晓媛
张治针
赵亚
吴俞莹
郑文婷
王政文
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zhejiang University ZJU
Original Assignee
Zhejiang University ZJU
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zhejiang University ZJU filed Critical Zhejiang University ZJU
Priority to CN202211636359.9A priority Critical patent/CN116019819A/en
Publication of CN116019819A publication Critical patent/CN116019819A/en
Pending legal-status Critical Current

Links

Images

Landscapes

  • Steroid Compounds (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

The invention provides an active ginsenoside composition, and a preparation method and application thereof, and belongs to the technical field of biological medicines. The invention utilizes the precious traditional Chinese medicine ginseng and American ginseng as well as the cheap traditional Chinese medicine ginseng stem and leaf, american ginseng stem and leaf, notoginseng stem and leaf and extracted total saponins to prepare the Active Total Ginsenoside Composition (ATGC), the Active Holographic Ginsenoside Composition (AHGC) and the active panaxadiol saponin composition (APDSC), which not only can save the ginseng traditional Chinese medicine, but also can greatly reduce the preparation cost of the product, and ensure the continuous supply of the raw materials for the subsequent industrial production of ATGC, AHGC and APDSC products; the complementarity of the active panaxadiol saponin monomer components in the stem and leaf and the active panaxadiol saponin monomer components of ginseng and American ginseng is utilized, so that the biological activity and pharmacological activity of ATGC, AHGC and APDSC and the health care value and the medicinal value thereof exceed the prior value.

Description

Active ginsenoside composition and preparation method and application thereof
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to an active ginsenoside composition, a preparation method and application thereof.
Background
For a long time, researches on biological activities and pharmacological actions of ginseng, american ginseng and pseudo-ginseng and Ginsenoside (ginsenosides) contained therein have been widely focused by scholars at home and abroad, and particularly, researches on actions and action mechanisms of single ginsenosides including panaxatriol saponins (such as GRg1, GRe and GRf) and panaxadiol saponins (such as GRb1, GRb2, GRb3, GRc and GRd) have been focused on by scholars at home and abroad. To date, thousands of reviews and research papers have focused on the biological activities of ginseng and ginsenoside, including central nervous system, cardiovascular system, immune system, endocrine system and tumor, and the main research results can be summarized as follows: 1) The ginseng and the ginsenoside have the functions of adapting to original shape (namely, the coping capability of the organism to various harmful stimuli from inside and outside the body can be improved, thereby slowing down or avoiding the harm of the harmful stimuli to the organism), regulating metabolism, resisting oxidation, resisting inflammation and resisting aging. Consistently, as a conceptual cognition, the effects of American ginseng and pseudo-ginseng, which also have ginsenoside as a main active ingredient, on the central nervous system, cardiovascular system, immune system, energy metabolism and adaptation as such are also widely accepted. 2) Panaxatriol saponin represented by GRg1 and panaxadiol saponin represented by GRb1 are widely studied and reported as antioxidants, anti-inflammatory agents and mitochondrial metabolism regulators, have the effect of protecting the nervous system and the cardiovascular system, panaxatriol saponin (GRg and GRe) and panaxadiol saponin (GRc) can also reduce insulin resistance (i.e. the body is insensitive to insulin), and inhibition of the p38 MAPK signal pathway is considered as one of the important mechanisms of action of panaxatriol saponin (GRg 1, GRe and GRf) and panaxadiol saponin (GRb 1, GRb3, GRc and GRd). Therefore, the panaxatriol saponins and panaxadiol saponins have been widely accepted as the main active ingredients of ginseng, american ginseng and pseudo-ginseng, and are considered to have wide medical application and development prospects. 3) The biological activities and pharmacological effects of panaxatriol saponins and panaxadiol saponins have the above-mentioned commonalities and also have heterogeneity differences, such as: panaxatriol saponin GRg1 is considered as a central stimulant, while panaxadiol saponin GRb is a central sedative, so that both components endow ginseng with activity of regulating the balance of central excitability and inhibition, and can cause the situation that the two components antagonize each other to impair the efficacy of the ginseng in treating related brain diseases, even causing side effects. In the same way, in practical application, the higher GRg-containing person participates in the American ginseng with GRb-containing higher efficacy with different emphasis, cannot replace each other, and is the applicable disease. 4) The biological activity and pharmacological effects of saponin components of the same type but different structures are common and have respective bias, such as: GRb2 it is believed that TGF- β1 and Smad signaling pathways are inhibited, SIRT1 expression is activated, and glucose metabolism is promoted, fat accumulation is reduced, epithelial-mesenchymal transition is inhibited, so that anticancer effect, myocardial inflammation and oxidative stress are reduced, and myocardial ischemia reperfusion injury is protected; GRb3 is believed to enhance myocardial function and immunity; GRc is believed to have antiallergic, antitumor, analgesic and sedative effects. In particular, the mechanisms by which ginsenosides produce the same pharmacological action may be different, e.g., different ginsenosides each potentiate the inhibitory activity of the central nerve by different mechanisms of action: activation of inhibitory GABA receptors (GRc and glycine receptors (GRb) and inhibition of excitatory aspartic acid (NMDA) receptors (GRg 3) and panaxatriol saponins such as GRg1 do not activate or act very poorly.
In conclusion, the diversity of the chemical structures of the ginsenosides and the similarity, diversity, heterogeneity and antagonism of the biological activity and pharmacological action of the ginsenosides reveal the modern scientific connotation of the efficacy and the drug properties of ginseng, american ginseng and pseudo-ginseng as traditional rare traditional Chinese medicines, simultaneously deepen the broad promotion of the body-building effect of the rare traditional Chinese medicines by people, and also initiates the interest and the expectation of professionals on researching the novel ginsenoside medicines to prevent and treat the chronic complex diseases such as brain diseases, cardiovascular diseases, immune dysfunction diseases, metabolic syndrome and the like. However, the medicinal value of the traditional rare traditional Chinese medicines is embodied in the form of total saponins of ginseng, and besides the single ginseng soup, the traditional rare traditional Chinese medicines are all in the form of compound administration of the traditional Chinese medicines. Although research and development of new drugs with single ginsenoside as an active ingredient have been widely paid attention, few patent people have been made at present. Obviously, a single ginsenoside cannot bear the functional connotation of the raw medicinal materials, and is less likely to effectively prevent and treat the complex diseases.
How to utilize the similarity, diversity and heterogeneity of the biological activity and pharmacological action of the ginsenoside to better exert the tonifying and body-building effects and important medicinal value of the rare traditional Chinese medicines is an important and complex scientific difficulty. Theoretically, the commonality, heterogeneity and reverse effects of biological activity and pharmacological effects exhibited by the different individual saponin components of panaxatriol saponin and panaxadiol saponin and its cognate saponins are both possible to integrate these active components to produce a completely new pharmaceutical form that encompasses and surpasses the efficacy of ginseng, american ginseng and notoginseng, but at the same time makes this integration process difficult and uncertain.
Disclosure of Invention
Accordingly, the present invention has been made in view of the above problems, and an object of the present invention is to provide an active ginsenoside composition, which can effectively prevent and treat various complex diseases by determining the active ingredients of a drug against various diseases based on the commonality, heterogeneity and reverse actions of the biological activities and pharmacological actions exhibited by different individual saponin components of panaxatriol saponins, panaxadiol saponins and the like.
The invention also aims to provide a preparation method of the active ginsenoside composition, which is configured to be used for preventing and treating various different diseases or playing health care functions according to the difference complementation characteristics of the content of single saponin components in the total saponins of different medicinal materials of the ginseng.
The invention also aims to provide an application of the active ginsenoside composition, fully and reasonably utilizes the unique biological activity and pharmacological action of each single ginsenoside and the synergistic action of the ginsenosides, and has great significance and application value in the aspects of medicine or health care.
The invention provides an active ginsenoside composition, which comprises a functional unit 1 consisting of GRb and GRd and a functional unit 2 consisting of GRc and GRb 3;
the mass ratio of the functional unit 1 consisting of GRb and GRd to the functional unit 2 consisting of GRc and GRb is 0.66-1.92.
Preferably, the active ginsenoside composition is an active panaxadiol saponin composition;
in the active panaxadiol saponin composition, the mass ratio of GRb to GRd is 0.79 to 2.08, the mass ratio of GRb to GRc is 0.67 to 2.17, the mass ratio of GRb1 to GRb3 is 0.82 to 2.76, and the mass ratio of GRc to GRb3 is 0.79 to 2.11;
preferably, the mass ratios of GRb, GRc, GRb3 and GRd include at least one of the following, but are not limited to: 2.17:1.00:1.27:2.12, 1.38:1.00:0.63:1.74, 1.00:1.00:0.47:0.64, 0.76:1.00:0.68:0.52, 0.75:1.00:0.56:0.66, 1.39:1.00:1.09:0.91 and 1.01:1.00:1.09:0.73; and allows the ratio of the components of each group to be varied within 10%;
further preferably, the active panaxadiol saponin composition further comprises GRb2;
the total mass percentage of the active panaxadiol saponin composition is more than 85% of GRb, GRc, GRb2, GRb3 and GRd, wherein the content of GRb2 is 0.1% -16%.
Preferably, the active ginsenoside composition is an active holographic ginsenoside composition or an active total ginsenoside composition;
the active holographic ginsenoside composition or active total ginsenoside composition comprises panaxatriol saponin and panaxadiol saponin; the panaxatriol saponins are mainly, but not limited to GRg and GRe; the panaxadiol saponins are mainly but not limited to GRb1, GRb3, GRc and GRd; wherein the ratio of the total mass of the panaxadiol saponins to the total mass of the panaxatriol saponins is 1.88-4.41, the mass ratio of GRe to GRg1 is 2.31-4.41, the mass ratio of GRb1 to GRe is 0.64-1.86, the mass ratio of GRb1 to GRd is 0.79-2.08, the mass ratio of GRb1 to GRc is 0.67-2.17, the mass ratio of GRb1 to GRb3 is 0.82-2.76, and the mass ratio of GRc to GRb3 is 0.79-2.11;
Preferably, the mass ratios of GRg, GRe, GRb1, GRc, GRb3 and GRd include at least one of the following, but are not limited to: 0.60:2.13:2.17:1.00:1.27:2.12, 0.76:2.16:1.38:1.00:0.63:1.74, 0.37:0.86:1.00:1.00:0.47:0.64, 0.28:0.66:0.76:1.00:0.68:0.52, 0.39:0.91:0.75:1.00:0.56:0.66, 0.23:0.91:1.39:1.00:1.09:0.91 and 0.21:0.76:1.01:1.00:1.09:0.73; and allows the ratio of the components of each group to be varied within 10%;
further preferably, the active holographic ginsenoside composition or active total ginsenoside composition further comprises GRb;
still more preferably, the total mass percentage of the active holographic ginsenoside composition is more than 70% of GRg, GRe, GRb1, GRc, GRb2, GRb and GRd, based on 100%, specifically comprising the following components in percentage: 3.22 to 7.71 percent of GRG1, 11.99 to 21.87 percent of GRe,12.62 to 19.82 percent of GRb1,8.42 to 18.82 percent of GRc,5.22 to 10.45 percent of GRb2,6.35 to 17.14 percent of GRb3 and 9.83 to 17.85 percent of GRd;
the total mass percent of the active total ginsenoside composition is more than 50 percent, calculated as 100 percent, of GRg1, GRe, GRb1, GRc, GRb2, GRb3 and GRd, and the active total ginsenoside composition specifically comprises the following components in percentage by weight: 2.12 to 5.91 percent of GRG1,9.05 to 16.77 percent of GRe,9.31 to 19.12 percent of GRb1,6.42 to 14.33 percent of GRc,3.58 to 7.96 percent of GRb2,4.87 to 12.94 percent of GRb3 and 7.28 to 13.60 percent of GRd, but the total content of each component is equal to or more than 100 percent.
The invention provides a preparation method of the active ginsenoside composition, which comprises the following steps:
dissolving total saponins of Ginseng radix, and loading into reversed phase C 18 In the silica gel chromatographic column, firstly, 43 volume percent of ethanol aqueous solution is used for eluting, when the ginsenoside GRb1 is detected, 50 to 55 volume percent of ethanol aqueous solution is used for eluting, and the eluent is collected until the ginsenoside GRd is not detected by the eluent, the elution is stopped, the collected eluent is combined and concentrated and dried by a conventional method to obtain an active panaxadiol saponin composition, or the fractions of the panaxadiol saponins GRb1, the GRc, the GRb2, the GRb3 and the GRd are detected on a combination line and are respectively collected, and then the active panaxadiol saponin composition is obtained by mixing according to specific mass; or dissolving total saponins of Ginseng radix in reverse phase C 18 In the silica gel chromatographic column, firstly, 30 percent by volume of ethanol aqueous solution is used for eluting, when the ginsenoside GRg1 is detected, 50 to 55 percent by volume of ethanol aqueous solution is used for eluting, and eluent is collected until the ginsenoside GRd is not detected by the eluent, the elution is stopped, the collected eluent is combined and concentrated and dried by a conventional method to obtain an active holographic ginsenoside composition, or the fractions of the ginsenoside GRg1, the GRe, the GRb1, the GRc, the GRb2, the GRb3 and the GRd are detected on a combination line and then mixed according to a specific mass to obtain the active holographic ginsenoside composition;
Or mixing total saponins of Ginseng radix as raw material to obtain active total ginsenoside composition.
Preferably, the solvent for dissolving the total saponins of the ginseng crude drug is ethanol water solution with the volume percentage content of 30 percent; the mass volume ratio of the total saponins of the ginseng crude drug to the 30% ethanol water solution is 1mg: 8-12 mL;
the method also comprises the following steps before loading: the reversed phase C is subjected to the process of adopting 30 percent ethanol water solution by volume percent 18 Performing balance treatment on the silica gel chromatographic column; the C is 18 The mass ratio of the usage amount of the silica gel to the total saponins of the raw medicinal materials is (7-10): 1.
preferably, the total saponins of the ginseng crude drug comprise the following combination: the composition comprises a combination of American ginseng root or American ginseng root total saponin and American ginseng stem and leaf or American ginseng stem and leaf total saponin, a combination of ginseng root or American ginseng root total saponin, ginseng stem and leaf or ginseng stem and leaf total saponin and American ginseng stem and leaf or American ginseng stem and leaf total saponin, a combination of ginseng root or ginseng root total saponin, ginseng stem and leaf or ginseng stem and leaf total saponin, and a combination of American ginseng root or American ginseng stem and leaf total saponin, american ginseng stem and leaf or American ginseng stem and leaf total saponin and notoginseng stem and leaf or American ginseng stem and leaf total saponin;
The mass ratio of the raw materials or total saponins of each ginseng in each combination is determined according to the content of each ginsenoside contained in each raw material or total saponin which is actually used;
preferably, in the combination of the American ginseng root total saponins and the American ginseng stem and leaf total saponins, the mass ratio of the American ginseng root total saponins to the American ginseng stem and leaf total saponins is 1 (2-3);
the mass ratio of the ginseng root total saponins, the American ginseng root total saponins, the ginseng stem and leaf total saponins and the American ginseng stem and leaf total saponins in the combination of the ginseng root total saponins, the American ginseng root total saponins, the ginseng stem and leaf total saponins and the American ginseng stem and leaf total saponins is 1:1:1:3, a step of;
preferably, in the combination of the ginseng root total saponins, the ginseng stem and leaf total saponins and the notoginseng stem and leaf total saponins, the mass ratio of the ginseng root total saponins, the ginseng stem and leaf total saponins and the notoginseng stem and leaf total saponins is (1-2): 1: (1-2);
in the combination of the American ginseng root total saponins, the American ginseng stem and leaf total saponins and the pseudo-ginseng stem and leaf total saponins, the mass ratio of the American ginseng root total saponins to the American ginseng stem and leaf total saponins is 1:1 (1-2);
further preferably, the total saponins of the ginseng crude drug are extracted from the ginseng crude drug or are commercial products;
The total saponins of the ginseng crude drug extracted from the ginseng crude drug are prepared by mixing the ginseng crude drug and then using a conventional method, or the total saponins of the crude drugs are prepared by a conventional method respectively, and then the total saponins of the ginseng crude drug are obtained after mixing; preferably, the extraction method comprises the steps of mixing the ginseng raw material, percolating and extracting for 3 times by using 70-90% ethanol water solution with the volume percentage content, and removing the solvent to obtain the ginseng raw material total saponins.
The invention provides application of the active ginsenoside composition or the active ginsenoside composition prepared by the preparation method in preparing medicines for preventing and/or treating diseases or health care products for exerting health care effects.
Preferably, the use of an active panaxadiol saponin composition and/or an active holographic panaxadiol saponin composition for the manufacture of a medicament for the prevention and/or treatment of a disease comprising at least one of the following: neurological disorders, autoimmune diseases, stress disorders and aging and related diseases;
preferably, the neurological condition comprises at least one of: mental disorders, sleep disorders, delayed and/or dysdevelopmental disorders, nerve injury and dysfunctional disorders, neurodegenerative disorders, addictive disorders and peripheral neuropathy;
Further preferably, the psychotic disorder comprises at least one of: schizophrenia, depression, bipolar disorder, anxiety and mania;
the sleep disorder includes at least one of: difficulty falling asleep, sleep maintenance disorder, early wake, reduced sleep quality, dreaminess, and reduced total sleep time; meanwhile, daytime dysfunction is accompanied, and daytime drowsiness, fatigue, inattention and hypomnesis occur, and tension, forcing and low emotion are accompanied;
the delayed and/or dysplastic condition comprises at least one of the following: hyperactivity, inattention, learning disorders, attention deficit hyperactivity disorder, autism, language disorders, sleep disorders, tourette syndrome, and tourette syndrome;
the nerve injury and dysfunctional disease includes at least one of: epilepsy, acute phase of cerebral apoplexy and cerebral apoplexy, neuropsychiatric dysfunction and akinesia sequela;
the neurodegenerative disease comprises at least one of: parkinsonism, alzheimer's disease, vascular dementia, chorea, multiple sclerosis and progressive freezing;
the parkinsonism comprises at least one of the following: multisystem degeneration-parkinsonism superposition, primary parkinsonism, juvenile parkinsonism, secondary parkinsonism caused by infection or ischemia, hereditary parkinsonism, and parkinsonism-like symptoms caused by drug therapy, i.e., extrapyramidal systemic responses; the multisystem degeneration-parkinsonism syndrome stack includes, but is not limited to, at least one of: multisystemic atrophy, progressive supranuclear palsy and corticobasal degeneration;
The addictive disorders include at least one of the following: alcohol and drug addiction, juvenile networking and gaming addiction, and pathological gambling;
the peripheral neuropathy includes at least one of: neuralgia, facial neuritis, facial spasm, multiple peripheral neuropathy, guillain-Barre syndrome, neuralgia and dyskinesia caused by viral infection;
preferably, the autoimmune disease comprises at least one of: lupus erythematosus, autoimmune glomerulonephritis, rheumatoid arthritis, dermatomyositis, scleroderma, allergic rhinitis, allergic asthma, urticaria, allergic conjunctivitis, demyelinating diseases, connective tissue diseases, neuromuscular diseases, digestive system diseases, endocrine diseases, and urinary system diseases;
further preferred, the connective tissue disease comprises at least one of: systemic lupus erythematosus, rheumatoid arthritis, dermatomyositis and scleroderma;
the neuromuscular disease includes at least one of the following: multiple sclerosis, myasthenia gravis, and demyelinating diseases;
the digestive system disease includes at least one of: chronic non-specific ulcerative colitis, chronic active hepatitis and pernicious anaemia and atrophic gastritis;
The endocrinopathy includes at least one of the following: primary adrenocortical atrophy and chronic thyroiditis;
the urinary system diseases comprise autoimmune glomerulonephritis and/or pulmonary renal hemorrhagic syndrome;
preferably, the stress disorder comprises chronic stress-induced sub-health status and/or stress wound sequelae;
further preferred, the sub-health status includes insomnia, dreaminess and daytime dysfunction and its associated daytime sleepiness, fatigue weakness, inattention, hypomnesis, reduced work efficiency and creativity, or with anxiety, obsessive-compulsive, depressed mood;
further preferred, the stress wound sequelae comprises at least one of: acute stress disorders, maladaptation and post-traumatic stress disorders;
preferably, the aging and its related diseases include at least one of the following: premature senility, senile hypomnesis, senile hypertension, senile sleep disorder, senile constipation and senile chronic inflammation.
Preferably, the dosage form of the medicament comprises a liquid formulation and/or a solid formulation;
the liquid preparation comprises oral liquid and/or injection;
the solid formulation includes, but is not limited to, at least one of the following: tablets, capsules, granules, pills, enteric-coated preparations, controlled-release preparations and nano-preparations.
Preferably, the application comprises the application of the active panaxadiol saponin composition or the active holographic ginsenoside composition as the only active ingredient and/or the synergistic attenuation of the active panaxadiol saponin composition or the active holographic ginsenoside composition combined with the preparation of the existing medicine to prepare a composite preparation. Preferably, the existing drug includes, but is not limited to, at least one of the following: levodopa and dopamine 2 receptor agonists for the treatment of parkinson's disease, dopamine receptor inhibitors for anti-schizophrenia and anxiety disorders, antiepileptic and neurological damage, sodium/calcium ion channel inhibitors for the treatment of neuropsychiatric behavioral disorders characterized by glutamate hyperexcitability or concomitant GABA-inhibitory deficiency, mycophenolate mofetil, antitumor chemotherapeutic or target agent for the treatment of autoimmune diseases and anti-organ transplant rejection;
further preferably, the levodopa drug comprises: meadouba, xiding and Daling complex;
dopamine 2 receptor agonists include, but are not limited to: arvensis (pramipexole dihydrochloride), ropinirole and cabergoline;
the inhibitors of the baamine receptor include, but are not limited to: haloperidol, olanzapine, clozapine, and risperidone;
The sodium and calcium ion channel inhibitors include, but are not limited to: lamotrigine or gabapentin;
when the medicinal preparation prepared from the active panaxadiol saponin composition and lamotrigine are used together or the compound medicament prepared from the active panaxadiol saponin composition and lamotrigine are used for exerting synergistic attenuation effect, the medicinal preparation is used for treating epilepsy, bipolar disorder and acute cerebral ischemia injury, and comprises but is not limited to: cerebral ischemia of birth canal of newborn in acute stage of cerebral apoplexy;
the medicinal preparation prepared by combining the active panaxadiol saponins and gabapentin are used or the composite medicament prepared by combining the active panaxadiol saponins and the gabapentin plays a role in synergism and attenuation and is used for treating hypoevolutism or development disorder, alcohol and medicament addiction, teenager network and game addiction and pathological gambling;
such chemotherapeutic or target agents include, but are not limited to: paclitaxel and tyrosine kinase inhibitors.
Preferably, the active total ginsenoside composition is applied to the preparation of health care products with health care functions, wherein the health care functions comprise at least one of the following components: delaying aging, improving sub-health status, improving health level and life quality of the elderly, and relieving nervous system side effects caused by drug treatment;
preferably, the drug treatment causes side effects of the nervous system including side effects of the nervous system caused by neuropsychiatric drugs or anti-tumor drugs;
Preferably, the neuropsychiatric drug comprises at least one of: dopamine receptor inhibitors and agonists, levodopa drugs, 5-hydroxytryptamine drugs, sodium-calcium channel inhibitors, glutamate receptor inhibitors, and other neurotransmitter receptor inhibitors;
the antitumor drug comprises at least one of the following: chemotherapeutic agents, molecular targeting agents, and immunotherapeutic agents;
preferably, the anti-aging comprises at least one of: inhibiting or preventing premature senility, improving senile hypomnesis, improving sleep, stimulating appetite, relieving senile chronic inflammation, improving senile mobility and prolonging life;
preferably, the sub-health state comprises at least one of the following states: insomnia, dreaminess, sleep disorders, tension, anxiety, depressed mood, hypomnesis, physical and mental fatigue, and reduced work efficiency.
Preferably, the health product comprises a liquid health care product and/or a solid health care product;
the liquid health care product comprises oral liquid;
the solid health care product comprises at least one of: tablets, capsules, granules and pills.
The invention provides an attenuated synergistic pharmaceutical composition, wherein the active ingredients of the pharmaceutical composition comprise an active panaxadiol saponin composition in the active panaxadiol saponin composition and/or an active holographic panaxadiol saponin composition in the active panaxadiol saponin composition, and the active pharmaceutical composition is obtained by combining at least one of the following pharmaceutical active ingredients: levodopa, dopamine type 2 receptor agonists, dopamine receptor inhibitors, chemotherapeutic or target agents, sodium and calcium channel inhibitors and mycophenolate mofetil.
Preferably, the inhibitor of the baamine receptor comprises at least one of the following: haloperidol, olanzapine, and risperidone;
the sodium and calcium ion channel inhibitors include lamotrigine or gabapentin.
The invention provides a large health product, which comprises an active total ginsenoside composition in the active ginsenoside composition or is prepared by combining at least one of the following components: nutritional ingredients and active ingredients;
the nutritional ingredients include at least one of the following: proteins, polypeptides and glutathione precursor amino acids, nad+ precursors and nucleic acids;
the active ingredients comprise at least one of the following: the medicine and food homology traditional Chinese medicine extract, coenzyme Q10, vitamins and energy metabolism intermediates.
Preferably, the large health product comprises an oral formulation;
the oral formulation comprises a solid formulation and/or a liquid formulation;
the solid formulations include, but are not limited to: capsules, normal tablets, dispersible tablets, enteric-coated tablets and granules.
The active ginsenoside composition provided by the invention comprises a functional unit 1 formed by GRb and GRd and a functional unit 2 formed by GRc and GRb 3; the mass ratio of the functional units 1 and GRc formed by GRb and GRd to the functional unit 2 formed by GRb and GRc is 0.66-1.92. According to the invention, through experiments of a total saponin active ingredient disassembly formula of ginseng traditional Chinese medicinal materials, the influence of the central excitability effect of panaxatriol saponin on the central inhibitory effect of panaxadiol saponin is clarified, and further analysis and verification prove that the content of panaxadiol saponin GRb2 obtained does not influence the efficacy of an active panaxadiol saponin composition, the panaxadiol saponins GRb, GRb3, GRc and GRd are determined to be main active ingredients of the efficacy of the panaxadiol saponin composition, and the four active ingredients form two functional units of 'GRb 1+ GRd' and 'GRc + GRb 3', and the two functional units respectively exert the unique effects of a direct implementation and a coordinator of the curative effect of different diseases or different diseases of the same disease, and the subtle synergistic effect (unavailability) of the two functions not only eliminates the respective efficacy defects, but also greatly improves the efficacy, safety and stability of the product and increases the effective dosage range. The invention enables GRb1, GRb3, GRc and GRd to respectively exert unique biological activity and pharmacological action through scientific compatibility of the ginsenoside, and simultaneously exert therapeutic value for effectively preventing and treating major diseases including neurodegenerative diseases through mutual synergistic action.
Further, the present invention is characterized in that the active ginsenoside composition is specifically classified into an Active Holographic Ginsenoside Composition (AHGC), an active panaxadiol saponin composition (APDSC) and an Active Total Ginsenoside Composition (ATGC) according to the variety and content of the active ingredients. The three compositions strictly define the ratio between the individual ginsenosides, thus further conferring the following technical features to the composition: (1) The biological activity and pharmacological activity of different ginsenosides in the ginseng traditional Chinese medicinal materials and the total saponins thereof are integrated, so that the ginseng total saponins have wide values of health care and disease prevention and treatment; (2) Avoiding the central excitatory action of panaxatriol saponin from weakening the central inhibitory action of panaxadiol saponin; (3) Avoiding the central excitation of panaxatriol saponin to possibly produce the effect of not conforming to the product function; (4) The optimized ratio of 7 ginsenosides provides various schemes for preparing target composition products by flexibly utilizing the mixing preparation of the raw materials of the Chinese medicinal materials of the ginseng, and can relieve the shortage of the resources of the Chinese medicinal materials of the ginseng and improve the use efficiency of the ginseng. ATGC and/or AHGC have the characteristics of strong function, stability, reliability and high safety. APDSC further excludes any direct side effect of central excitability of triol saponin and pharmacological action of weakening diol saponin in use scene, thus endowing APDSC with clinical localization characteristics more suitable for preventing and treating nervous system diseases characterized by glutamate excitability and gamma-aminobutyric acid inhibition deficiency than AHGC. In addition, the 7 preferred ratios of the quality standards of ATGC and AHGC and the 7 preferred ratios of the quality standards of APDSC provide various schemes for flexibly utilizing the available ginseng medicinal materials to prepare target products, thereby relieving the shortage of ginseng medicinal material resources and improving the use efficiency of the ginseng medicinal materials.
Further, the invention specifically defines that the active ginsenoside composition further comprises GRb2.GRb2 as an ingredient which is not related or relevant to the medical and health care effects, allows it to exist in natural state in Active Holographic Ginsenoside Composition (AHGC), active panaxadiol saponin composition (APDSC) and Active Total Ginsenoside Composition (ATGC) products.
The invention provides a preparation method of the active ginsenoside composition, which takes Chinese medicinal materials of ginseng as raw materials and takes alcohol aqueous solution as an extraction solvent to obtain living organismsThe total ginsenoside composition is prepared by mixing total saponins of Chinese medicinal materials of Panax, and is subjected to reversed phase C 18 Separating with silica gel chromatographic column. The preparation method has the advantages of easily available raw materials and good repeatability, and lays a foundation for clinical drug development and preparation of health care effect products.
Drawings
FIG. 1 HPLC chromatogram of total ginsenoside in example 1.
FIG. 2 is an HPLC chromatogram of total saponins of stems and leaves of ginseng in example 1.
FIG. 3 HPLC chromatogram of total saponins of American ginseng root in example 1.
FIG. 4 HPLC chromatogram of total saponins of stems and leaves of American ginseng in example 1.
FIG. 5 HPLC chromatogram of total saponins of stem and leaf of Notoginseng in example 1.
FIG. 6 is an HPLC chromatogram of the ginsenoside composition PDSC1 in example 2.
Fig. 7 is an HPLC chromatogram of the panaxatriol saponin composition PTSC1 of example 2.
FIG. 8 is an HPLC chromatogram of holographic ginsenoside composition HGC1 in example 4.
FIG. 9 is an HPLC chromatogram of holographic ginsenoside composition HGC2 in example 4.
FIG. 10 is an HPLC chromatogram of holographic ginsenoside composition HGC3 in example 4.
FIG. 11 is an HPLC chromatogram of holographic ginsenoside composition HGC4 in example 4.
FIG. 12 HPLC chromatogram of holographic ginsenoside composition HGC5 in example 4.
FIG. 13 is an HPLC chromatogram of holographic ginsenoside composition HGC6 in example 4.
FIG. 14 is an HPLC chromatogram of holographic ginsenoside composition HGC7 in example 4.
FIG. 15 is an HPLC chromatogram of holographic ginsenoside composition HGC8 in example 4.
FIG. 16 is an HPLC chromatogram of holographic ginsenoside composition HGC9 in example 4.
FIG. 17 HPLC chromatogram of holographic ginsenoside composition HGC10 in example 4.
FIG. 18 is an HPLC chromatogram of the ginsenoside composition PDSC1 of example 6-1.
FIG. 19 is an HPLC chromatogram of the ginsenoside composition PDSC2 of example 6-1.
FIG. 20 is an HPLC chromatogram of the ginsenoside composition PDSC4 of example 6-1.
FIG. 21 is an HPLC chromatogram of the ginsenoside composition PDSC5 of example 6-1.
FIG. 22 is an HPLC chromatogram of the ginsenoside composition PDSC6 of example 6-1.
FIG. 23 is an HPLC chromatogram of the ginsenoside composition PDSC7 of example 6-1.
FIG. 24 is an HPLC chromatogram of the ginsenoside composition PDSC9 of example 6-1.
FIG. 25 HPLC chromatogram of the ginsenoside composition PDSC10 of example 6-1.
FIG. 26. Effect of ROT on p-p65 levels in mouse microvascular endothelial cells (bEnd.3), primary mouse Astrocytes (ASC) and Microglial Cells (MCG), dopamine neuron PC12 cells in example 10-2.
FIG. 27 effects of ROT on the pathological processes of neurovascular units and alpha-synuclein and PDSC1 during the development and onset of the rat model PD in examples 10-6.
FIG. 28 effects of ROT and PDSC1 on Striatum (Striatum) and substantia nigra pars compacta (SNc) and GFAP, NF-B and iNOS thereof in examples 10-6.
FIG. 29 protective effect of PDSC1 on vascular lesions and peripheral astrocytes in the striatum and substantia nigra brain areas of PD animals in examples 10-6 and maintenance of drug efficacy of PDSC1 in combination with L-DOPA.
FIG. 30 protection of PDSC1 from damage by microvascular and blood brain barrier in the striatum and dense parts and ventral parts of the brain of PD animals in examples 10-6.
FIG. 31 ROT-induced reduction of PD rat striatal PV positive neurons and protection of PDSC1 in example 11.
Fig. 32 neuroprotective effect of PDSC1 in example 16 against global cerebral ischemia in mongolian gerbils. A: typical changes in the CA1 region of the hippocampus 7 days after ischemia are shown for each experimental group; b: the number of neurons (mean ± SEM) that survived or degenerated over a length of 250 μm in the CA1 region of each group is shown. ++ p <0.001vs. sham surgery control group; * p <0.05, < p <0.01vs. model control group; group #p <0.05vs. MK-801.
FIG. 33, hippocampal nerve injury profile and neuroprotection by PDSC1 over time following ischemia in example 16. Mean number of neurons Surviving (SEM) in the middle region of CA1, 250 μm long, was shown at each time point p <0.01vs. model control.
Fig. 34. Neuroprotective effects of PDSC1 on whole brain ischemia in mongolian gerbil in example 16. The average number of surviving or degenerating neurons (mean±sem) in the central region of CA1, 250 μm long, is shown. ++ p <0.001vs. sham surgery control group; * p <0.05vs. model control group; group #p <0.05vs. MK-801.
FIG. 35. Protection of PDSC1 and neurological deficit induced by angiotensin ET-1 in example 16 following focal ischemia in rats. Shown in the figure are mean ± SEM of behavior scores for each experimental group; * p <0.05, < p <0.001vs. model group.
FIG. 36 neuroprotection of ET-1 induced focal ischemia in rats by PDSC1 in example 16. A: typical histological changes of cortex and hippocampus; b: lesion volumes of cortex and striatum; ++ p <0.01vs. sham surgery group; * p <0.05, < p <0.01vs. model group.
FIG. 37 combination of PDSC1 with lamotrigine in example 17-1 resulted in excellent antiepileptic efficacy in isoniazid and thiocarbamide induced acute epileptic models.
FIG. 38 shows that PDSC1 alone and in combination with LTG in example 17-2 has good therapeutic effect on pilocarpine-induced refractory chronic epilepsy in rats.
FIG. 39. Adverse effects of PDSC1 in combination with LTG in examples 17-4 can combat LTG-induced dermatitis in mice.
FIG. 40A is a graph showing the development of butterfly-like erythema in the cheek of a pristane-induced Systemic Lupus Erythematosus (SLE) model mouse in example 18.
Fig. 41 comparative graph of treatment of butterfly erythema in cheek for 60 days of administration to each group of mice in example 18.
Detailed Description
The invention provides an active ginsenoside composition, which comprises a functional unit 1 consisting of GRb and GRd and a functional unit 2 consisting of GRc and GRb 3; the mass ratio of the functional unit 1 consisting of GRb and GRd to the functional unit 2 consisting of GRc and GRb is 0.66-1.92.
In the present invention, the active ginsenoside composition preferably includes an active panaxadiol saponin composition (APDSC), an Active Holographic Ginsenoside Composition (AHGC), and an Active Total Ginsenoside Composition (ATGC) according to the ratio of active ingredients and the difference in kinds of active ingredients. The experimental screening of the invention determines that four ginsenosides GRb1, GRb3, GRc and GRd are main active ingredients of the active ginsenoside composition, and the four active ingredients form two functional units of GRb < 1 > + GRd > and GRc + GRb3 < 3 >. The two functional units exert the wonderful actions of a direct practitioner and a coordinator of the curative effects of the two functional units on different diseases or different symptoms of the same disease respectively, and the subtle synergistic action (unappropriately) of the two functional units not only eliminates the respective drug effect defects, but also greatly improves the drug effect, the stability and the safety of the product and increases the range of effective dosages. In particular, the active panaxadiol saponin composition (APDSC) product further completely eliminates any side effects that the central excitability effect of panaxatriol saponin weakens the central inhibitory effect of panaxadiol saponin under the use scene, which gives the active panaxadiol saponin composition (APDSC) product more suitable for preventing and treating clinical localization characteristics of nervous system diseases characterized by glutamate hyperexcitability and gamma-aminobutyric acid insufficient inhibitory effect than the active holographic panaxadiol saponin composition (AHGC) product. Examples of motor symptoms of parkinsonism alleviation experimental results prove that in the composition, the functional unit 1 formed by GRb and GRd is a direct drug effect implementation, but the effective dosage range is narrow, the dose-effect relationship shows jumping bidirectional change, the functional unit 2 formed by GRc and GRb3 is a coordinator, the functional unit 2 formed by GRc and GRb3 and the functional unit 1 formed by GRb1 and GRd cooperatively exert positive drug effects, and side effects of the functional unit 1 formed by GRb1 and GRd can be restrained, so that the overall drug effect is remarkable, stable and reliable. In contrast, experimental results of examples of efficacy against the occurrence and development of parkinson's disease demonstrate that the functional unit 2 is a direct effector of efficacy, but its effective dosage range is narrow and even punctiform, while functional unit 1 develops or shows weak protective efficacy depending on its dosage or exacerbation of PD, but can cooperate with functional unit 2 to produce a stronger efficacy and can greatly expand the effective dosage range.
In the invention, the mass ratio of GRb1 to GRd is 0.79 to 2.08, the mass ratio of GRb1 to GRc is 0.67 to 2.17, the mass ratio of GRb1 to GRb3 is 0.82 to 2.76, and the mass ratio of GRc to GRb3 is 0.79 to 2.11; the mass ratio of (GRb 1+ GRd)/(GRc + GRb 3) is preferably 0.66 to 1.92. The mass ratios of GRb, GRc, GRb3 and GRd preferably include at least one of the following ratios: 2.17:1.00:1.27:2.12, 1.38:1.00:0.63:1.74, 1.00:1.00:0.47:0.64, 0.76:1.00:0.68:0.52, 0.75:1.00:0.56:0.66, 1.39:1.00:1.09:0.91 and 1.01:1.00:1.09:0.73. the ratio of 6 is used as the basis of the preferable quality standard of the active panaxadiol saponin composition and the mixing and preparing material proportion of the traditional Chinese medicinal materials or the total saponin raw materials used for preparing the active panaxadiol saponin composition. In addition, the seven preferable ratio of GRb/GRc/GRb 3/GRd provides a plurality of schemes for preparing the target composition product by flexibly utilizing the mixed preparation of the ginseng traditional Chinese medicine raw materials, and can relieve the shortage of the ginseng traditional Chinese medicine resources and improve the use efficiency.
In the present invention, the active panaxadiol saponin composition preferably further comprises GRb. GRb2 as an ingredient which is not related or relevant to the pharmaceutical and health care effects described, allows it to be present in the active panaxadiol saponin composition product in a natural state. The total mass percentage of the panaxadiol saponins in the active panaxadiol saponins composition is preferably more than 85%, more preferably more than 90%, based on 100%, and the active panaxadiol saponins composition specifically comprises the following components in percentage by weight: 19.93 to 28.14 percent of GRb1, 12.95 to 26.30 percent of GRc,7.32 to 15.32 percent of GRb2, 10.61 to 23.88 percent of GRb3 and 13.73 to 29.47 percent of GRd. In the present invention, the active holographic ginsenoside composition or active total ginsenoside composition contains the same active ingredient, except that the ratio of active ingredients is different, preferably comprising panaxatriol saponin and panaxadiol saponin. The panaxatriol saponins are preferably GRg1 and GRe; the panaxadiol saponins are preferably GRb1, GRb3, GRc and GRd. Wherein the active holographic ginsenoside composition meets the 8 ratio standard, preferably, the ratio of the total mass of the panaxadiol saponins to the total mass of the panaxatriol saponins is 1.88-4.41, the mass ratio of GRe to GRg1 is 2.31-4.41, the mass ratio of GRb1 to GRe is 0.64-1.86, the mass ratio of GRb1 to GRd is 0.79-2.08, the mass ratio of GRb1 to GRc is 0.67-2.17, the mass ratio of GRb1 to GRb3 is 0.82-2.76, and the mass ratio of GRc to GRb3 is 0.79-2.11; the mass ratio of GRg, GRe, GRb1, GRc, GRb3 and GRd comprises 0.60:2.13:2.17:1.00:1.27:2.12, 0.76:2.16:1.38:1.00:0.63:1.74, 0.37:0.86:1.00:1.00:0.47:0.64, 0.28:0.66:0.76:1.00:0.68:0.52, 0.39:0.91:0.75:1.00:0.56:0.66, 0.23:0.91:1.39:1.00:1.09:0.91 and 0.21:0.76:1.01:1.00:1.09:0.73. the seven ratios GRg/GRe/GRb 1/GRc/GRb3/GRd are independent of each other, namely, each preferable ratio represents a composition product formed by GRg, GRe, GRb1, GRc, GRb3 and GRd according to the specific proportion, wherein the proportion of the panaxatriol saponin and the panaxadiol saponin contained and the proportion of each single panaxadiol saponin are restrained, thus further endowing the composition product with the following technical characteristics: (1) Integrates the biological activity and pharmacological activity of different ginsenosides in the ginseng traditional Chinese medicinal materials and the total saponins thereof, thereby having wide values of health care and disease prevention and treatment; (2) Avoiding the central excitatory action of panaxatriol saponin from weakening the central inhibitory action of panaxadiol saponin; (3) Avoiding the central excitation of panaxatriol saponin to possibly produce the effect of not conforming to the product function; (4) The seven preferred ratios provide a plurality of schemes for preparing the target composition product by flexibly utilizing the mixing and preparation of the ginseng traditional Chinese medicine raw materials, and can relieve the shortage of ginseng traditional Chinese medicine resources and improve the use efficiency.
In the invention, the structural formulas of GRg and GRe are shown as structural formula I:
Figure BDA0004007353830000081
wherein GRg represents ginsenoside Rg1 (r=h), GRe represents ginsenoside Re (r=6-deoxy-1-methylpyranoyl).
The structural formulas of GRb, GRc, GRb3 and GRd are shown in structural formula II:
Figure BDA0004007353830000082
wherein GRb represents ginsenoside Rb1 (R=β -D-glucopyranosyl), GRc represents ginsenoside Rc (R=α -L-arabinofuranosyl), GRb2 represents ginsenoside Rb2 (R=α -L-arabinopyranosyl), GRb3 represents ginsenoside Rb3 (R=β -D-xylopyranosyl), GRd represents ginsenoside Rd (R=H).
In the present invention, the active holographic ginsenoside composition or active total ginsenoside composition preferably further comprises GRb2.GRb2 as an ingredient which is not related or relevant to the pharmaceutical and health care effects described, allows it to be present in natural state in Active Holographic Ginsenoside Composition (AHGC) and Active Total Ginsenoside Composition (ATGC) products.
In the present invention, the total mass percentage of the active holographic ginsenoside composition is more than 70%, more preferably more than 80% of GRg, GRe 1, GRc, GRe 2, GRb3 and GRd, based on 100%, specifically comprising the following components in percentage: 3.22 to 7.71 percent of GRG1, 11.99 to 21.87 percent of GRe,12.62 to 19.82 percent of GRb1,8.42 to 18.82 percent of GRc,5.22 to 10.45 percent of GRb2,6.35 to 17.14 percent of GRb3 and 9.83 to 17.85 percent of GRd.
In the present invention, the total mass percentage of the GRg, GRe 1, GRc, GRe 2, GRb3 and GRd is 50% or more, more preferably 60% or more, based on 100% of the active total ginsenoside composition, and specifically comprises the following components in percentage: 2.12 to 5.91 percent of GRG1,9.05 to 16.77 percent of GRe,9.31 to 19.12 percent of GRb1,6.42 to 14.33 percent of GRc,3.58 to 7.96 percent of GRb2,4.87 to 12.94 percent of GRb3 and 7.28 to 13.60 percent of GRd. The invention provides a preparation method of an active ginsenoside composition, which specifically comprises a preparation method of an active panaxadiol saponin composition or an active holographic ginsenoside composition and a preparation method of an active total ginsenoside composition.
The preparation method of the active panaxadiol saponin composition or the active holographic panaxadiol saponin composition comprises the following steps:
dissolving total saponins of Ginseng radix, and loading into reversed phase C 18 Eluting with 43% ethanol water solution by volume percentage in silica gel chromatographic column, eluting with 50-55% ethanol water solution by volume percentage when detecting ginsenoside GRb1, collecting eluate until ginsenoside GRd is not detected by eluate, concentrating and drying the combined eluate to obtain active panaxadiol saponin composition.
Or dissolving total saponins of Ginseng radix in reverse phase C 18 Eluting with 30% ethanol water solution by volume percentage in silica gel chromatographic column, eluting with 50-55% ethanol water solution by volume percentage when detecting ginsenoside GRg1, collecting eluate until ginsenoside GRd is not detected by eluate, concentrating and drying the combined eluate to obtain active holographic ginsenoside composition.
In the invention, based on the content difference of single ginsenoside at different parts of each Chinese medicinal material of ginseng, the active ginsenoside composition is prepared by taking specific application as guiding mixing preparation. The invention respectively takes American ginseng root, american ginseng stem and leaf, ginseng root, ginseng stem and leaf, notoginseng stem and leaf or the corresponding total saponins as raw materials to prepare the active ginsenoside composition. The total saponins of the ginseng crude drug preferably comprise the following combination: the composition comprises the combination of American ginseng root total saponins and American ginseng stem and leaf total saponins, the combination of ginseng root total saponins, american ginseng root total saponins, ginseng stem and leaf total saponins and American ginseng stem and leaf total saponins, the combination of ginseng root total saponins, ginseng stem and leaf total saponins and pseudo-ginseng stem and leaf total saponins, and the combination of American ginseng root total saponins, american ginseng stem and leaf total saponins and pseudo-ginseng stem and leaf total saponins. Wherein, in the combination of the American ginseng root total saponins and the American ginseng stem and leaf total saponins, the mass ratio of the American ginseng root total saponins to the American ginseng stem and leaf total saponins is preferably 1 (2-3). In the combination of the ginseng root total saponins, the American ginseng root total saponins, the ginseng stem and leaf total saponins and the American ginseng stem and leaf total saponins, the mass ratio of the ginseng root total saponins, the American ginseng root total saponins, the ginseng stem and leaf total saponins and the American ginseng stem and leaf total saponins is preferably 1:1:1:3. in the combination of the ginseng root total saponins, the ginseng stem and leaf total saponins and the notoginseng stem and leaf total saponins, the mass ratio of the ginseng root total saponins, the ginseng stem and leaf total saponins and the notoginseng stem and leaf total saponins is preferably (1-2): 1: (1-2), and the ratio of the total saponins of the ginseng root is not lower than that of the total saponins of the stem and leaf of the pseudo-ginseng. In the combination of the American ginseng root total saponins, the American ginseng stem and leaf total saponins and the pseudo-ginseng stem and leaf total saponins, the mass ratio of the American ginseng root total saponins, the American ginseng stem and leaf total saponins and the pseudo-ginseng stem and leaf total saponins is preferably 1:1 (1-2). The source of the total saponins of the ginseng crude drug is not limited, and the total saponins can be purchased from a commercialized way or extracted from the ginseng crude drug. The total saponins of the ginseng crude drug extracted from the ginseng crude drug are prepared by mixing the ginseng crude drug and then using a conventional method, or the total saponins of the crude drugs are prepared by a conventional method respectively, and then the total saponins of the ginseng crude drug are obtained after mixing; preferably, the extraction method comprises the steps of mixing the ginseng raw material, percolating and extracting the ginseng raw material with 70-90% ethanol water solution for 3 times, and removing the solvent to obtain the ginseng raw material total saponins. The method for removing the solvent is not particularly limited, and the method for removing the solvent well known in the art may be used. The mass volume ratio of the ginseng crude drug to the 70-90% ethanol aqueous solution by volume percent is 1mg:10 to 15mL, more preferably 1mg:12mL. In the embodiment of the invention, the American ginseng root total saponins, the American ginseng stem and leaf total saponins, the ginseng root total saponins, the ginseng stem and leaf total saponins and the pseudo-ginseng stem and leaf total saponins are all commercial products.
At the bookIn the invention, the solvent for dissolving the total saponins of the ginseng crude drug is preferably ethanol water solution with the volume percentage content of 30 percent; the mass volume ratio of the total saponins of the ginseng crude drug to the 30% ethanol water solution is preferably 1mg:8 to 12mL, more preferably 1mg:10mL. The reverse phase C before the loading 18 The silica gel chromatographic column is subjected to equilibrium treatment by adopting 30% ethanol water solution by volume percent. Ginseng crude drug and reversed phase C 18 The mass ratio of the silica gel is preferably 1:7-10. The opposite phase C 18 The silica gel chromatographic column is preferably of the specification of COSMIL 75C 18 PREP filler.
In the present invention, in preparing the active panaxadiol saponin composition, elution is preferably performed using 43% by volume of ethanol aqueous solution. During elution, the flow rate is preferably 150-200 mL/min, the eluent is collected, each component in the eluent is analyzed by HPLC, the elution by 43% ethanol water solution is stopped until ginsenoside GRb1 appears in the component, then the elution is continued by 55% ethanol water solution, each component is obtained by collecting 55% ethanol eluent, and the elution by 55% ethanol water solution is stopped until ginsenoside GRd does not appear in the component; finally, the components containing GRb, GRc, GRb2, GRb3 and GRd are combined, concentrated under reduced pressure to be completely dried to obtain an active panaxadiol saponin composition (APDSC), and the respective contents of five ginsenosides in the active panaxadiol saponin composition (APDSC) are measured by an HPLC method, and the relative proportion of the contents and the total content of the five ginsenosides are calculated.
In the present invention, in preparing the active holographic ginsenoside composition, elution is preferably performed by using 30% ethanol aqueous solution by volume percent. In the case of elution, the flow rate is preferably 150 to 200mL/min. Collecting eluent while eluting, analyzing each component in the eluted solution by High Performance Liquid Chromatography (HPLC), stopping eluting by 30% ethanol water solution until ginsenoside GRg1 appears in the component, then continuing eluting by 55% ethanol water solution, collecting eluent of 55% ethanol water solution to obtain each component, and analyzing each component by HPLC until ginsenoside GRd does not appear in the component, stopping eluting by 55% ethanol water solution; finally, the eluents containing GRg, GRe 1, GRe 2, GRb and GRd were combined, concentrated under reduced pressure to complete dryness to obtain an Active Holographic Ginsenoside Composition (AHGC), and the respective contents of seven ginsenosides in the Active Holographic Ginsenoside Composition (AHGC) were measured by HPLC method, and the relative proportion of the contents and total content of seven ginsenosides were calculated.
In the preparation method of the active total ginsenoside composition, the active total ginsenoside composition is obtained by mixing total saponins of the ginseng crude drug. The mixing scheme of total saponins of ginseng crude drugs is the same as the mixing configuration scheme of total saponins in the preparation of active holographic ginsenoside composition, and is not described herein.
The invention provides application of the active ginsenoside composition or the active ginsenoside composition prepared by the preparation method in preparing medicines for preventing and/or treating diseases or health care products for exerting health care effects.
In the present invention, ATGC, AHGC and APDSC integrate the body-strengthening and body-consolidating effects and the biological activity or pharmacological actions of the adaptation of ginseng, and 4 kinds of panaxadiol saponins (including Rb1, rc, rb3 and Rd) and their relative content configurations are cores for supporting the functions of the product, and can regulate the homeostasis including energy metabolism homeostasis, redox balance homeostasis and neuroendocrine immune homeostasis and improve the adaptive homeostasis ability of the body, thus slowing down or even avoiding the loss of physical and mental health reserves due to the in vitro and in vivo harmful pressures or events (also called stressors, including physical, chemical/pharmaceutical therapeutic, biological and mental harmful pressure sources), causing sub-health states, causing diseases and accelerating the aging process; the state of the neuroendocrine immune system which has deviated from the physiological range can also be regulated back to the physiological range, thereby alleviating the symptoms of the related diseases and promoting the self-healing of the diseases. Therefore, ATGC has great application value in promoting great health, and simultaneously AHGC and/or APDSC have wide medicinal value.
In the present invention, the development of broad pharmaceutical value by AHGC and/or APDSC is achieved based on a common mechanism. The results of the examples of the present invention demonstrate that the common mechanisms of AHGC and/or APDSC treatment of various diseases includeThe following contents are: AHGC and/or APDSC are capable of protecting and repairing functional and structural homeostasis of individual member cells in the central nervous vascular unit and peripheral congeneric functional units (hereinafter collectively referred to as functional units) to maintain and reestablish microenvironment homeostasis (including but not limited to energy, redox, NAD + Neurotransmitter), thereby, can resist or relieve the disease risk factors from inside and outside the body or/and pathogenic events from injuring fragile members in the functional units to cause pathological changes of other members and pathological vicious circle among subsequent members, and can slow down or even cut off the pathological vicious circle among each member in the units and repair the deficiency and damage, finally achieve the purpose of maintaining and repairing the functions and the structural homeostasis of micro blood vessels, glial cells, neurons and nerve fibers, and realize the medical value for preventing and treating the nervous system diseases and delaying the aging, which is characterized by treating both symptoms and root causes.
In the present invention, the use of a holographic ginsenoside composition and/or a panaxadiol saponin composition for the preparation of a medicament for the prevention and/or treatment of a disease comprising at least one of the following: nervous system disorders, autoimmune diseases, stress diseases and aging and related diseases.
In the present invention, the neurological disorder preferably includes at least one of: mental disorders, developmental delay and dysdevelopmental disorders, neurological injuries and dysfunctional disorders, neurodegenerative disorders, addictive disorders and peripheral neurodegenerative disorders. The psychotic disorder preferably comprises at least one of: schizophrenia, depression, bipolar disorder, anxiety and mania. The bradykinesia and dysplastic conditions preferably include at least one of the following: hyperactivity, inattention, learning disorders, attention deficit hyperactivity disorder, autism, language disorders, sleep disorders, tourette syndrome, and tourette syndrome. The nerve injury and dysfunctional disorder preferably includes at least one of: epilepsy, cerebral apoplexy, neuropsychiatric dysfunction and akinesia sequela. The neurodegenerative disease preferably comprises at least one of the following: parkinson's disease, alzheimer's disease, vascular dementia, chorea, multiple sclerosis and progressive freezing. The addictive disorder preferably comprises at least one of the following: alcohol and drug addiction, juvenile networking and gaming addiction, and pathological gambling. The peripheral neurodegenerative disease preferably comprises at least one of the following: neuralgia, facial neuritis, facial spasm, multiple peripheral neuropathy, guillain-Barre syndrome, neuralgia caused by viral infection and dyskinesia.
In the embodiment of the invention, the application of AHGC or APDSC in preventing and treating mental diseases. Mental diseases are characterized by hyperparathyroidism (Glu), insufficient gamma-aminobutyric acid (GABA) inhibitory signals, and hyperparathyroidism (DA). The main functions of basal ganglia include motor control, reinforcement learning, emotion motivation and other advanced cognitive functions, the striatum is the core of basal ganglia, and the functions of the basal ganglia are mainly received from excitatory input taking Glu as transmitter from cerebral cortex and thalamus, DA input of dopamine nerve of substantia nigra compact part and regulation and control of GABA transmitter of striatum interneurons, and after mutual integration, the signals of the transmitters realize regulation and control of cognition, emotion, motor function and habitual behavior through output of the spiny neurons. The hyperexcitation of striatal Glu, insufficient GABA inhibitory signals and the hyperactivation of DA signals lead to psychotic disorders including schizophrenia, while GABA inhibitory interneuron (PV-Ins) disorders are also important pathological features of the above-mentioned neuropsychiatric disorders. Furthermore, astrocyte atrophy and loss of its function are important features of most neuropsychiatric dysfunctional diseases, being the pathophysiological cause of schizophrenia, major Depressive Disorder (MDD) and bipolar disorder (BPD). Astrocytes are critical for maintaining normal levels of extracellular Glu and GABA, while astrocyte atrophy and loss of function must result in hyperactivity of its Glu, further exacerbating the excitatory and inhibitory imbalance caused by GABA function. Furthermore, psychotic disorders are associated with disorders of the intestinal microbiota. Because the AHGC and APDSC compositions containing the two functional units "GRb1+ GRd" and "GRc + GRb3" protect astrocytes and inhibitory interneurons (PV-Ins) from damage and normal function, maintain or even reconstruct Glu excitability and GABA inhibitory balance or neurochemical homeostasis in the striatum and other brain regions, protect healthy homeostasis of the intestinal flora, to protect against the key links of the disease network. Therefore, AHGC and/or APDSC have application value and potential in the prevention of the neuropsychiatric dysfunctional diseases.
In the embodiment of the invention, the application of AHGC or APDSC in preventing and treating epilepsy is realized. The mechanism of preventing and treating epilepsy by AHGC or APDSC containing two functional units of 'GRb & lt1+ & gt GRd' and 'GRc & lt GRb3 & gt' is as follows: (1) Isoniazid and thiocarbamide which are synthesis inhibitors of p-aminobutyric acid (GABA) can obviously induce acute epileptic seizure of rats, and the combined use of the isoniazid and thiocarbamide with the prior antiepileptic drug lamotrigine can greatly improve the antiepileptic drug effect, and is obviously shown as further reducing the severity of epileptic behavior and reducing the number of animals with large seizure; (2) The AHGC or APDSC and the lamotrigine are used singly or in combination to correct the hyperexcitability of the chronic epileptic brain by increasing GABA and reducing glutamic acid so as to return to a physiological or near physiological central inhibitory and excitatory dynamic balance state, so that the combined use of the AHGC and the APDSC and the lamotrigine not only can overcome the serious defect of epileptic seizure rebound after the lamotrigine drug withdrawal, but also can generate sustainable anti-epileptic drug efficacy, and has excellent drug efficacy that the efficacy is quick in onset, does not resist drugs, and does not appear drug withdrawal rebound but can be continued; (3) The combination of AHGC or APDSC with lamotrigine can completely treat serious side effects of mice dermatitis, neuropsychiatric disorders (such as challenge aggression, anxiety, confusion, hallucinations, ataxia) and adverse reactions of extrapyramidal systems (such as the occurrence of parkinsonism-like or chorea-like actions) caused by epilepsy with lamotrigine; the use of AHGC and APDSC, alone or in combination with the antiepileptic drug lamotrigine, has application value and potential in the treatment of acute and chronic epilepsy and in reducing the side effects of lamotrigine.
In the embodiment of the invention, the application of AHGC and/or APDSC in preventing and treating cerebral apoplexy (apoplexy) is realized. Since mitochondrial dysfunction, oxidative stress injury, inflammatory response are intimately associated with acute stroke and chronic brain injury, disruption of neurovascular units and impaired function, including cerebrovascular endothelial cells, astrocytes and neural cells, are common pathological features of stroke. AHGC and/or APDSC containing the two functional units of GRb & lt 1 & gt GRd & lt GRc & lt GRb & gt can protect cerebral vascular endothelial cells, astrocytes and nerve cells from damage and functional damage, maintain ATP steady state to resist mitochondrial dysfunction and oxidative stress damage and inflammatory reaction caused by the mitochondrial dysfunction, prevent epileptic attacks in the acute phase of cerebral ischemia injury of animals, quickly restore the motor function damaged in the acute phase of ischemia reperfusion, reduce the death rate in the acute phase and protect neurons in the ischemic brain region. Therefore, AHGC and/or APDSC have application value and potential for preventing and treating cerebral apoplexy.
In the embodiment of the invention, the Active Holographic Ginsenoside Composition (AHGC) or the active panaxadiol saponin composition (APDSC) shows good drug effect in preventing and treating the Parkinson Disease (PD), and two functional units of 'GRb 1+ GRd' and 'GRc + GRb 3' in the AHGC or the APDSC can maintain and protect the energy metabolism steady state, the oxidation reduction steady state, the mitochondrial function and the oxidation coenzyme I (NAD) + ) And Glutathione (GSH) and reduced coenzyme ii (NADPH) -mediated physiological functions, promote the regeneration of NADPH and GSH from their oxidized forms and increase GSH synthesis to enhance the ability of endothelial cells with deficient mitochondrial complex enzyme I function to scavenge Reactive Oxygen Species (ROS), protect neurovascular units including cerebrovascular endothelial cells, astrocytes and nerve cells, and inhibit the formation of neurotoxic phenotype A1 cells, reestablish the neurochemical homeostasis of glutamate (Glu) and aminobutyric acid (GABA) in the striatum of animals, maintain central excitability and inhibitory balance, and have a uniformly significant therapeutic effect in AHGC or APDSC on rat parkinson's disease induced by inhibition of dopamine receptor-induced mice stiffness symptoms reflecting the palliative effect of the test drug on parkinson's disease and by inhibition of mitochondrial complex enzyme I-induced rotenone reflecting the essential effect of the test drug on parkinson's disease. And the combined administration of AHGC or APDSC and the existing medicament for treating parkinsonism, namely L-DOPA, can remarkably improve the curative effect of the L-DOPA, reduce the catastrophe caused by the treatment of the L-DOPA, and prevent the schizophrenia such as reduced learning ability, mental nerve behavior disorder, delusions and hallucinations caused by the treatment of the L-DOPA. In addition, changes in intestinal microbiota, damage to intestinal epithelial barriers, Intestinal inflammation and neuroplastic rearrangement of the enteric nervous system are associated with the pathophysiology of PD intestinal disorders. While AHGC or APDSC can protect the intestinal flora against the toxicity of PD risk factor pesticides present in the environment or against mitochondrial dysfunction in vivo, it is also the mechanism of action of AHGC or APDSC to prevent PD from developing and developing. In conclusion, the AHGC or APDSC can be used for treating various symptoms of the Parkinson Disease (PD) and can delay the progress of the disease, improve the curative effect of the L-DOPA and reduce the serious toxic and side effects caused by the treatment of the L-DOPA medicament.
In the examples of the present invention, since Alzheimer's disease, vascular dementia, chorea, multiple sclerosis and progressive freezing have the same pathological characteristics, neurovascular unit destruction and functional impairment including cerebrovascular endothelial cells, astrocytes and nerve cells, mitochondrial dysfunction is accompanied by insufficient Adenosine Triphosphate (ATP) energy, oxidative stress injury and neuroinflammation, etc., these pathological characteristics are closely related to the occurrence and development of diseases. The two functional units of GRb1+ GRd and GRc + GRb3 contained in the AHGC or APDSC can protect the damage and functional damage of cerebrovascular endothelial cells, astrocytes and nerve cells, maintain and protect mitochondrial functions, maintain the energy substance ATP at a sufficient level, prevent and treat oxidative stress injury, inhibit NF-B pathway and the release of adhesion factors and inflammatory factors, and the AHGC or APDSC can prolong the life of premature animals and remarkably improve learning and memory capacity. Similarly, changes in intestinal microbiota, damage to intestinal epithelial barriers and intestinal inflammation are closely related to gastrointestinal diseases such as alzheimer's disease, vascular dementia, chorea, multiple sclerosis and progressive freezing, and AHGC or APDSC can protect the patient's intestinal microbiota homeostasis and inhibit inflammatory response. Therefore, AHGC or APDSC has application value and potential in preventing and treating Alzheimer's disease, vascular dementia, chorea, multiple sclerosis and progressive freezing disease.
In embodiments of the invention, AHGC and/or APDSC are used in the prevention and treatment of addictive disorders. The deficiency of dopamine type 2 receptors (D2 receptors) results in individuals having a variety of addictive, impulsive and compulsive behaviors, such as alcohol addiction, various drug and drug addiction, juvenile game addiction, pathological gambling, chronic violence and antisocial behaviors. In agreement, alcohol and almost all drugs and drugs are met by patients by increasing dopamine release, whereas long-term dopamine overdose further weakens the otherwise low D2R receptor function and the overall dopaminergic circuit changes, resulting in more intense dependence and withdrawal symptoms for the patient on alcohol, drugs and drugs. In addition, the deficiency of aminobutyric acid (GABA) inhibitory function and glutamate (Glu) hyperexcitation directly mediate withdrawal symptoms and are also closely related to game addiction. There are two main classes of drugs currently in use or in clinical trials to combat the various addiction: (1) Acamprosate (a non-specific GABA receptor agonist) and topiramate (activating GABA type a receptor) that promote GABA function; (2) Topiramate, lamotrigine and gabapentin (inhibiting presynaptic voltage-gated sodium and calcium channels and thus inhibiting Glu release) inhibit Glu excitatory signals. AHGC and/or APDSC products containing two functional units, "GRb1+ GRd" and "GRc + GRb3", can be directed against the effects of the D2 receptor inhibitor haloperidol, as well as against the effects of chronic treatment of levodopa that overstimulates D2 receptors resulting in some of their functions being desensitized and others being hypersensitive, and can bring back to near physiological levels too high Glu and GABA levels and too low GABA levels in the striatum. As can be seen, AHGC and APDSC can be fully regulated against neurochemical abnormalities in the addictive liability, during drug withdrawal and withdrawal phases, with the unique advantage of altering brain homeostasis back to normal (correcting D2 receptor insufficiency, insufficient GABA inhibition and Glu hyperexcitability in or towards normal in individuals with addictive liability and addicts, especially in withdrawal phases). Thus, AHGC and APDSC can provide new types of drugs for the prevention and treatment of alcohol addiction, drug and drug addiction, juvenile game addiction, pathological gambling addiction, sexual addiction, and various compulsive behaviors.
In the embodiment of the invention, the application of AHGC and/or APDSC in preventing and treating peripheral nerve disease. Although the causes of degenerative diseases of peripheral neuropathy are diverse, mitochondrial dysfunction is a major pathological mechanism of peripheral neuropathy, and in particular, mitochondrial dysfunction-mediated Adenosine Triphosphate (ATP) energy deficiency, oxidative stress, and inflammatory responses are all involved in the initiation and progression of peripheral neuropathy. The most commonly used drugs for the current treatment of peripheral neuropathy are the anticonvulsants gabapentin and pregabalin, which act by blocking presynaptic terminal voltage-gated sodium and calcium channels while down regulating the excitatory neurotransmitter glutamate (Glu) release, and in addition, vitamin B1, adenosylcobalamin and mecobalamin, which are believed to be useful in the treatment of peripheral neuropathy caused by various causes, are also used. Compared with the prior art and the prior art, the AHGC and/or APDSC containing two functional units of 'GRb & lt1+ & gt GRd' and 'GRc & lt GRb3 & gt' has unique action and action mechanism: namely, the initiation and development of diseases are prevented against the core pathological mechanism of peripheral neuropathy by protecting mitochondrial function, maintaining the homeostasis of energy metabolism and redox balance, preventing the occurrence of neuroinflammation, reducing Glu excitotoxicity and enhancing gamma-aminobutyric acid (GABA) inhibitory function. Therefore, AHGC and/or APDSC have special advantages and application values in the aspect of controlling the peripheral neuropathy.
In the present invention, the autoimmune disease preferably includes at least one of: allergic rhinitis, allergic asthma, urticaria, allergic conjunctivitis and autoimmune diseases caused by excessive immunity. The autoimmune disease caused by the excessive immunity preferably includes at least one of the following: connective tissue diseases, neuromuscular diseases, digestive system diseases, endocrine diseases and urinary system diseases. The connective tissue disease preferably comprises at least one of the following: systemic lupus erythematosus, rheumatoid arthritis, dermatomyositis and scleroderma. The neuromuscular disease preferably includes at least one of the following: multiple sclerosis, myasthenia gravis, and demyelinating diseases. The digestive system diseases preferably include at least one of the following: chronic nonspecific ulcerative colitis, chronic active hepatitis, pernicious anemia and atrophic gastritis. The endocrinopathy preferably includes at least one of the following: primary adrenocortical atrophy and chronic thyroiditis. The urinary system disease preferably comprises autoimmune glomerulonephritis and/or pulmonary renal hemorrhagic syndrome.
In the embodiment of the invention, the AHGC and/or APDSC products containing the two functional units of GRb1+ GRd and GRc + GRb3 can obviously inhibit butterfly-shaped erythema and reduce urine protein level on the cheek of a Systemic Lupus Erythematosus (SLE) mouse induced by an immunopotentiator pristane, and can completely inhibit butterfly-shaped erythema on the cheek of the SLE mouse when being combined with mycophenolate mofetil medicines, so that the AHGC and/or APDSC products are used singly or combined with mycophenolate mofetil and have advantages and application values in treating systemic lupus erythematosus diseases.
In the present invention, the stress-induced disease preferably includes chronic stress-induced related disease and/or stress wound sequelae. The chronic stress-induced related diseases preferably include at least one of: cardiovascular disease, diseases of the reproductive and endocrine system, diseases of the nervous system and diseases of the digestive system. The stress wound sequelae preferably comprises at least one of the following: acute stress disorder, maladaptation and post-traumatic stress disorder.
In embodiments of the present invention, the biological system is enabled for continuous short-term adjustments in constantly changing internal and external environments due to adaptive homeostasis to achieve optimal function. Reduced adaptive homeostasis can impair resistance to a variety of stressors (e.g., hypoxia, oxidative stress, immune response and mental stress), which when the intensity and duration of stress responses exceeds the stress sustained by the adaptive homeostasis function of the body can cause stress-related diseases (e.g., primary hypertension of negative mood-initiated and aggravated, acute stress-related responses, delayed stress-related responses and maladaptation, inflammatory-induced heart disease and autoimmune disease, immunosuppressed-induced viral infections and vaccination responses). Whereas intracellular availability of Adenosine Triphosphate (ATP) and oxidized coenzyme I (NAD) + ) Level and suitable NAD + NADH ratio is critical to maintaining cell adaptive homeostasis, ATP and NAD + Depletion can impair the adaptive stress response of cells, and ATP depletion can lead to depletion of reduced coenzyme ii (NADPH) and Glutathione (GSH) and inflammatory responses. Due to the inclusion of "GRb1+ GRd" and "GRc +"GRb3 "AHGC and/or APDSC of two functional units does not disrupt adaptive homeostasis of cells, but rather the self-rescue mechanism for selectively activating endothelial cells facing mitochondrial complex I insufficiency involves moderately increasing NAD + Level and NAD + the/NADH ratio (maintains ATP at a sufficient level), promotes regeneration of NADPH and GSH from its oxidized form, and increases GSH synthesis (increases the ability to scavenge ROS), increasing the ability of endothelial cells to cope with otherwise deleterious stressors. Therefore, the results of the embodiment of the invention support that AHGC and APDSC have great medical application value in the aspect of preventing and curing the stress diseases.
In the present invention, the aging and related diseases preferably include at least one of the following: senile constipation, hypertension, sleep disorders and chronic inflammation.
In the present invention, the dosage form of the drug preferably includes a liquid formulation and/or a solid formulation. The liquid preparation preferably comprises oral liquid and/or injection. The solid formulation preferably comprises at least one of the following dosage forms: tablets, capsules, granules, pills, enteric-coated preparations, controlled-release preparations and nano-preparations.
In the invention, the active total ginsenoside composition is preferably applied to the preparation of health care products with health care effects.
In the present invention, functions of ATGC include improving metabolic and nutritional status and neuroendocrine immune status, improving sleep, antioxidant, regulating intestinal flora, improving central growth and development, preventing premature senility and delaying aging, so ATGC can help a wide population to maintain good health and vitality, and can serve specific populations, including: the young and young sub-health syndrome/physical and mental exhaustion (such as physical weakness, reduced mental work efficiency, anxiety and depression, etc.), and sleep disturbance, people in long-term pressure in the heart society, people in high three highs, people with insufficient immune system activity or autoimmune diseases (or strain diseases, such as allergy and asthma, rheumatoid arthritis and lupus), patients who are and will face anti-tumor treatment toxicity, people facing radiation hazard, people who are long-term receiving hormone drugs (including glucocorticoids) or psychotic drugs, children and young people with neuropsychiatric retardation and disorder, people with heart disease and hypertension, men and women with climacteric symptoms, people with skin diseases, people recovering from chronic diseases, people with premature aging, and aged people with functional degradation (including but not limited to constipation, anorexia, hypomnesis, hypoimmunity, slow movement, insomnia).
In the present invention, the health care efficacy preferably includes at least one of: drug treatment causes side effects of the nervous system, delays aging and improves sub-health status.
In the present invention, the drug treatment causes side effects of the nervous system including side effects of the nervous system caused by neuropsychiatric drugs or antitumor drugs. The neuropsychiatric drug preferably comprises at least one of the following: dopamine receptor inhibitors and agonists, L-DOPA (L-DOPA), 5-hydroxytryptamine drugs, inhibitors of sodium-calcium ion channels, glutamate receptor inhibitors, and other neurotransmitter receptor inhibitors. The antitumor drug preferably comprises at least one of the following: chemotherapeutic agents, molecular targeting agents, and immunotherapeutic agents.
In the embodiment of the invention, levodopa is used for treating parkinsonism, so that abnormal symptoms are often caused, and meanwhile, the patients can also have schizophrenia symptoms such as reduced learning ability, psychoneurosis, delusions and hallucinations. And the combined use of AHGC or APDSC and the levodopa can remarkably improve the curative effect of the levodopa, reduce the catastrophe caused by the treatment of the levodopa, and prevent the schizophrenia such as the reduced learning ability, the mental nerve behavior disorder, the delusions and the hallucinations caused by the treatment of the levodopa.
In the embodiment of the invention, the prior antiepileptic drug lamotrigine treats epilepsy and cannot avoid the number of animals with large seizures; the AHGC or APDSC and the lamotrigine are used singly or in combination, so that the serious defect of epileptic seizure rebound after lamotrigine drug withdrawal can be overcome, sustainable antiepileptic drug efficacy can be generated, and the medicine has the excellent efficacy of quick acting, no drug resistance, no drug withdrawal rebound and continuous drug efficacy; in addition, the use of AHGC or APDSC in combination with lamotrigine can completely treat serious side effects of mice dermatitis, neuropsychiatric disorders (e.g., challenge aggression, anxiety, confusion, hallucinations, ataxia) and adverse reactions of extrapyramidal systems (e.g., the appearance of parkinsonism-like or chorea-like actions) caused by epilepsy with lamotrigine.
In the present invention, the aging delaying preferably includes at least one of: improving sleep, stimulating appetite, improving activity and prolonging life.
In the examples of the present invention, oxidized coenzyme I (NAD + ) Deletions lead to mitochondrial dysfunction, adenosine Triphosphate (ATP) deficiency and Reactive Oxygen Species (ROS) accumulation, ultimately leading to high oxidative stress and cell senescence and death, while maintaining or restoring intracellular ATP and NAD + Level and NAD + NADH ratio is a strategy that is more effective in delaying senescence than one that is antioxidant and anti-inflammatory or targets a functional molecule of the mitochondria in the physiological range. AHGC and/or APDSC containing "GRb1+ GRd" and "GRc + GRb3" functional units can maintain and protect energy metabolic homeostasis, redox homeostasis, mitochondrial function and NAD + Glutathione (GSH) and reduced coenzyme II (NADPH) mediated normal physiological functions, prolongs the service life of the SAMP8 mice, and remarkably improves the learning and memory capacity of the SAMP8 mice and prevents the animal learning and memory capacity reduction caused by chronic treatment of levodopa, so that the AHGC and the APDSC have application value and potential in the aspect of delaying aging.
In the present invention, the sub-health status preferably includes at least one of the following status: stress, sleep disorders, anxiety, depression, fear, hypomnesis and physical and mental fatigue.
In embodiments of the present invention, ATGC achieves health care efficacy by improving sub-health status. Cell adaptive homeostasis enables continuous short-term regulation of biological systems in constantly changing internal and external environments to achieve optimal function. Reduced adaptive homeostasis can impair resistance to a variety of stressors, which when the intensity and duration of the stress-induced stress exceeds the stress sustained by the adaptive homeostasis function of the body can cause a variety of sub-health states including emotional stress, sleep disorders, anxiety, agitation, irritability, depression, hypomnesis, physical and mental fatigue, and reduced work efficiency. Adenosine Triphosphate (ATP) and oxidized coenzyme I (nad+) levels and appropriate nad+/NADH ratios are critical for maintaining cell adaptive homeostasis, ATP and nad+ depletion can impair cell adaptive stress responses, and ATP depletion can lead to reduced coenzyme ii (NADPH) and Glutathione (GSH) depletion and inflammatory responses. In addition, the weakness of the intestinal flora and dysregulation of the intestinal flora are also one of the important causes of sub-health. Since the ATGC containing the two functional units "GRb1+ GRd" and "GRc + GRb3" does not disrupt the adaptive homeostasis of the cell, but rather selectively activates endothelial cells that face a deficiency in mitochondrial complex I function, a self-rescue mechanism that includes moderately increasing nad+ levels and nad+/NADH ratios (maintaining ATP at sufficient levels), promoting regeneration of NADPH and GSH from their oxidized forms, and increasing GSH synthesis (increasing ROS scavenging capacity), increases the ability of endothelial cells to cope with otherwise deleterious stressors, and may protect the healthy homeostasis of the intestinal flora. Therefore, the results of the present examples support the health care use of the ATGC in improving the sub-health status.
In the present invention, the health care product preferably comprises a liquid health care product and/or a solid health care product. The liquid health care product preferably comprises an oral liquid. The solid health care product preferably comprises at least one of the following: tablets, capsules, granules and pills.
The invention provides an attenuated synergistic pharmaceutical composition, wherein the pharmaceutical active ingredients comprise an active panaxadiol saponin composition and/or an active holographic panaxadiol saponin composition in the active panaxadiol saponin composition, and the active panaxadiol saponin composition is combined with at least one of the following pharmaceutical active ingredients: meadob (mass ratio of levodopa to benserazide of 4:1), tannin (ratio of carbidopa to levodopa of 1:4), daltefacompound (levodopa 50 mg: carbidopa 12.5 mg: entacapone 200 mg), dopamine 2 receptor agonists, dopamine receptor inhibitors, chemotherapeutics or targets, sodium and calcium ion channel inhibitors and mycophenolate mofetil.
In the present invention, the inhibitor of the monoamine receptor preferably comprises at least one of the following components: haloperidol, olanzapine, and risperidone. The sodium and calcium ion channel inhibitors preferably comprise lamotrigine or gabapentin. The medicine prepared from the active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition is clinically combined with western medicines such as a baamine receptor inhibitor, so that the treatment effect can be effectively improved, the defect that the western medicines cure the symptoms but not the root cause (can not correct the disease) can be overcome, and the side effects of the western medicines can be relieved. In order to prove a specific treatment scheme after the active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition are combined with western medicines, the following technical scheme is specifically proposed.
The invention provides application of the pharmaceutical composition in preparing medicines for treating Parkinson's disease and/or relieving side effects caused by treating Parkinson's disease by levodopa, wherein the pharmaceutical composition comprises the levodopa and at least one of the following compositions: an active panaxadiol saponin composition and an active holographic panaxadiol saponin composition. The active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition are combined with the levodopa drugs to prepare the compound preparation, so that the effects of synergism and toxicity reduction can be effectively achieved. In the medicine, AHGC or APDSC plays roles of maintaining or reconstructing striatal non-dopamine neurotransmitter steady state and dopamine receptor function steady state, protecting nerve vascular units, and levodopa is used for supplementing missing dopamine but simultaneously disturbing dopamine receptor function steady state and non-dopamine transmitter steady state, particularly, the level of excitatory transmitter glutamic acid is rapidly and continuously increased, and the level of inhibitory transmitter gamma-aminobutyric acid is rapidly and continuously reduced, and the two agents are combined for treating Parkinson's disease, so that the striatal chemical steady state close to normal people can be reconstructed, and therefore, the movement and non-movement symptoms can be relieved, the disease progression is delayed and even blocked, and the side effects of catabolism, mental disorder and cognitive disorder caused by chronic treatment of levodopa are slowed and even eliminated.
The invention provides application of the pharmaceutical composition in preparing medicines for preventing and treating addictive diseases, wherein the pharmaceutical composition comprises a dopamine 2 receptor agonist and at least one of the following compositions: an active panaxadiol saponin composition and an active holographic panaxadiol saponin composition. The active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition are combined with the dopamine 2 receptor agonist to prepare the compound preparation, so that the synergistic and toxicity-reducing effects can be effectively achieved. In the medicine, the dopamine 2 receptor agonist combined with the active panaxadiol saponin composition or the active holographic panaxadiol saponin composition can effectively correct the weak brain (seeking stimulation tendency) and the gamma-aminobutyric acid inhibitory signals and dopamine signal deficiency in the addictive brain, thereby achieving the purpose of preventing and treating the addictive diseases.
The invention provides application of the pharmaceutical composition in preparing medicines for treating schizophrenia, wherein the pharmaceutical composition comprises a dopamine receptor inhibitor and at least one of the following compositions: an active panaxadiol saponin composition and an active holographic panaxadiol saponin composition. The active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition are combined with the dopamine receptor inhibitor to prepare the compound preparation, so that the synergistic and toxicity-reducing effects can be effectively achieved. In the medicine, the dopamine receptor inhibitor can alleviate the positive symptoms caused by dopamine 2 type receptor through inhibiting dopamine 2 type receptor, but the action of inhibiting dopamine 2 type receptor leads to extrapyramidal reaction, namely PD-like movement symptoms, and AHGC or APDSC can not only compensate the defect that the dopamine receptor inhibitor has no effect of relieving negative symptoms, but also can prevent extrapyramidal reaction, so that the two kinds of drugs can be combined to achieve the effect of treating schizophrenia.
The invention provides application of the pharmaceutical composition in preparing medicines for treating tumors, wherein the pharmaceutical composition comprises a chemotherapeutic drug or a target drug and at least one of the following compositions: an active panaxadiol saponin composition and an active holographic panaxadiol saponin composition. The active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition are combined with chemotherapeutics or target agents to prepare the compound preparation, so that the effects of synergism and toxicity reduction can be effectively achieved. In the medicine, the chemotherapeutic medicine or the target medicine still plays a role in killing tumors, and the AHGC or the APDSC can relieve negative psychology caused by fear of a patient on diseases and also can relieve cardiotoxicity, neurotoxicity, immunotoxicity and fatigue of the anti-tumor medicine, so that the combined treatment of the two can achieve the effects of attenuation and synergism on the tumors. The present invention provides the use of said pharmaceutical composition comprising lamotrigine and at least one of the following compositions for the preparation of a medicament for the treatment of epilepsy, bipolar disorders and conditions characterized by hyperactivity of Glu or simultaneous lack of GABA inhibitory activity: an active panaxadiol saponin composition and an active holographic panaxadiol saponin composition. The active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition are combined with lamotrigine to prepare a compound preparation, so that the effects of synergism and toxicity reduction can be effectively achieved. In the medicine, lamotrigine exerts curative effect by inhibiting Glu release, but simultaneously inhibits GABA release, and chronic treatment can trigger cells to express more ion channels so as to lead to gradual decrease of medicine effect and rebound of withdrawal symptoms, and common central side effects and severe dermatitis; AHGC or APDSC can integrate the effect of inhibiting Glu release by lamotrigine and establish a new stable balance between nerve excitability and inhibition, so that the treatment effect is stronger and more stable, and the side effect of LTG is avoided, and therefore, the two can be combined for use, and the toxicity-reducing and efficacy-enhancing medicine effect can be achieved.
The invention provides application of the pharmaceutical composition in preparing medicines for treating peripheral neuropathy or addictive diseases, wherein the pharmaceutical composition comprises gabapentin and at least one of the following compositions: an active panaxadiol saponin composition and an active holographic panaxadiol saponin composition. The active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition and the gabapentin are used for preparing a compound preparation, so that the effects of synergism and toxicity reduction can be effectively achieved.
The invention provides application of the pharmaceutical composition in preparing medicines for preventing and treating autoimmune diseases, skin diseases or rejection after liver and kidney transplantation, wherein the pharmaceutical composition comprises mycophenolate mofetil and at least one of the following compositions: an active panaxadiol saponin composition and an active holographic panaxadiol saponin composition. The compound preparation is prepared from the active panaxadiol saponin composition and/or the active holographic panaxadiol saponin composition and mycophenolate mofetil, so that the effects of synergism and toxicity reduction can be effectively achieved.
In the present invention, the dosage form of the drug preferably includes oral preparations and injection preparations. The oral formulation preferably comprises a solid and/or liquid formulation. The solid formulation preferably comprises at least one of the following dosage forms: capsules, ordinary tablets, dispersible tablets, enteric-coated tablets, granules, pills, enteric-coated preparations, controlled release preparations, and capsules containing micropellets or minitablets. The method for preparing the medicine is not particularly limited, and the method for preparing the medicine known in the art can be adopted. The taking method of the medicine comprises the step of taking the AHGC or the APDSC and the medicine active ingredient or the step of preparing the composite preparation from the AHGC or the APDSC and the medicine active ingredient. The dosage (time/d) of the AHGC or the APDSC in the medicament is preferably 25 mg-200 mg, more preferably 50 mg-100 mg, and the medicament active ingredient is 60% -100% of the dosage according to the clinical doctor's advice. In the composite preparation, the mass ratio of the AHGC or APDSC to the active pharmaceutical ingredients is 0.01-100: 1, more preferably 0.1 to 10:1.
The invention provides a large health product, which comprises an active total ginsenoside composition in the active ginsenoside composition or an oral product prepared by combining at least one of the following components: nutritional ingredients and active ingredients. The nutritional ingredients include at least one of the following: proteins, polypeptides and glutathione precursor amino acids, NAD + Precursors and nucleic acids. The active ingredients comprise at least one of the following: the Chinese medicine extract or active ingredient with homology of medicine and food, coenzyme Q10, vitamin and energy metabolism intermediate product.
In the present invention, the large health product preferably includes an oral formulation. The oral formulations include solid and/or liquid formulations. The solid formulations include, but are not limited to: capsules, normal tablets, dispersible tablets, enteric-coated tablets and granules. The preparation method of the large health product is not particularly limited, and the preparation method of the health product known in the art can be adopted. In the said health product, the weight of the active total ginsenoside composition in one dose is preferably 25 mg-200 mg, more preferably 50 mg-100 mg. The weight of the nutritional ingredients and active ingredients in a dose of product is preferably the usual dose of each substance as a nutraceutical.
The following examples are provided to illustrate the active ginsenoside composition, the preparation method and the application thereof in detail, but they should not be construed as limiting the scope of the invention.
Example 1
The contents of main panaxatriol saponins and panaxadiol saponins from ginseng root and its stem and leaf, american ginseng root and its stem and leaf, and notoginseng stem and leaf have a high complementary relationship.
The main active ingredients of the Chinese medicinal materials from the ginseng (Panax) plant comprise ginseng root and stem leaf, american ginseng root and stem leaf, notoginseng and notoginseng stem leaf, which are all various ginsenosides, but each of the Chinese medicinal materials is rich in the respective advantageous ginsenosides, and is also lack (low content, < 5%) or lack (is not detected under experimental conditions) other certain ginsenosides. Therefore, the comprehensive utilization of these medicinal materials can prepare a holographic ginsenoside composition (holographic ginsenoside combination, HGC) with biological activity and pharmacological action superior to those of the ginsenoside composition derived from the single medicinal material. Accordingly, the contents of panaxatriol saponins Rg1 (GRg 1) and Re (GRe) and panaxadiol saponins Rb1 (GRb 1), rb2 (GRb 2), rb3 (GRb 3), rc (GRc) and Rd (GRd) in commercial ginseng root total saponins, ginseng stem and leaf total saponins, american ginseng root total saponins, american ginseng stem and leaf total saponins prepared from raw medicinal materials derived in 2020 were measured by a High Performance Liquid Chromatography (HPLC) method provided under the Chinese medicinal ginseng item of the pharmacopoeia of the people's republic of China (part, page 9, 2020). In order to further refine the individual characteristics of the main ginsenosides contained in each medicinal material, the relative content of panaxatriol saponins and panaxadiol saponins and the relative content between different individual components were analyzed.
The results of the HPLC analysis are shown in tables 1 and 2 and FIGS. 1 to 5.
Table 1. The contents of main panaxatriol saponins and panaxadiol saponins in total saponins of various medicinal materials of Panax genus (%, n=3)
Figure BDA0004007353830000161
The total saponins of Ginseng radix comprise 11.97% of total saponins of panaxatriol (GRe/GRg 1=2.43) and 52.92% of total saponins of panaxadiol (GRb) 1 /GRc/GRb 2 /GRb 3 GRd =1.12/1.00/0.78/0.17/0.58), the high content (10-19%) of saponins has GRb (16.18%), GRc (14.50%) and GRb2 (11.36%), the medium content (5-9%) of saponins has GRe (8.48%) and GRd (8.39%), and the low content [ (-8.39%)<5%) of the saponins were GRe (3.49%) and GRd (2.49%).
The total saponins of Ginseng radix stem and leaf comprise 34.86% of total saponins of panaxatriol (GRe/GRg 1=2.41) and 32.32% of total saponins of panaxadiol (GRb) 1 /GRc/GRb 2 /GRb 3 GRd =1.38/1.00/1.17/0.35/3.26), GRe (24.62%) for very high content (. Gtoreq.20%) of saponins, GRd (14.71%) and GRg (10.23) for high content (10-19%) of saponins, GRb1 (6.21%) and GRb2 (5.28%) for medium content (5-9%) of saponins, low content [ ]<5%) of the saponins were GRc (4.51%) and Gb3 (1.59%).
The total saponins of radix Panacis Quinquefolii comprise 15.21% of total saponins of panaxatriol (GRe/GRg 1=6.96) and 59.10% of total saponins of panaxadiol (GRb) 1 /GRc/GRb 2 /GRb 3 GRd =3.26/1.00/0.34/0.33/1.12), GRb (31.95%) of very high content (. Gtoreq.20%) of saponins, GRe (13.30%) and GRd (11.01%) of high content (10-19%) of saponins, GRc (9.81%) of medium content (5-9%) of saponins, low content% <5%) of the saponins were GRb (3.38%), GRb3 (3.28%) and GRg1 (1.91%).
The total saponins of stems and leaves of American ginseng comprise 22.46% of total saponins of panaxatriol (GRe/GRg 1=2.74) and 40.16% of total saponins of panaxadiol (GRb) 1 /GRc/GRb 2 /GRb 3 According to/GRd =1.07/1.00/1.52/1.93/3.82), the saponins with high content (10-19%) have GRe (16.45%) and GRd (16.42%), the saponins with medium content (5-9%) have GRb (8.32%), GRb2 (6.52%) and GRg1 (6.00%), and the saponins with low content are [ - ]<5%) of the saponins were GRb (4.60%) and GRc (4.30%).
The total saponins of stem and leaf of Notoginseng radix contains 46.78% of total saponins of Ginseng radix glycol (GRb) 1 /GRc/GRb 2 /GRb 3 GRd =0.26/1.00/0.26/1.28/0.17), GRb (20.15%) of very high content (. Gtoreq.20%) of saponins, GRc (15.72%) of high content (10-19%) of saponins, low content%<5%) of the saponins were GRb (4.13%), GRb2 (4.08%) and GRd (2.71%), lacking panaxatriol saponins GRg1 and GRe.
TABLE 2 complementary relationship of ginsenoside as main active ingredient between different medicinal materials of Panax
Figure BDA0004007353830000162
Figure BDA0004007353830000171
The analysis results show that the contents of the main 7 ginsenosides contained in each medicinal material and the relative proportions of the contents are greatly different. Undoubtedly, the content characteristics of the 7 ginsenosides analyzed above support the efficacy and the existing clinical application of each medicinal material, and also suggest that each component of the ginsenosides derived from the medicinal materials may not effectively exert the full biological activity and pharmacological action of panaxatriol saponin and panaxadiol saponin and the related health care and medical application thereof. However, the individual components of ginsenosides derived from ginseng root, ginseng stem and leaf, american ginseng root, american ginseng stem and leaf and notoginseng stem and leaf have a good complementary relationship, and a Holographic Ginsenoside Composition (HGC) enriched with active components GRg, GRe, GRb1, GRc, GRb2, GRb3 and GRd can be prepared by using such a complementary relationship.
Ginsenoside GRg, GRe, GRb1, GRc, GRb2, GRb and GRd have wide biological activities of regulating metabolism, neuroendocrine and immunity, neuroprotection and the like, and also have wide pharmacological effects of resisting oxidation, resisting inflammation, and protecting organism cells such as nerves, cardiac myocytes, vascular endothelial cells and the like from being damaged by internal and external pathogenic factors. Thus, the active ginsenosides are integrated together to form a composition containing the main panaxatriol saponin and panaxadiol saponin active ingredients, which may be in a form that contains and exceeds the health care and medical value of ginseng root and its stem and leaf, american ginseng root and its stem and leaf, and notoginseng stem and leaf.
Accordingly, the present invention contemplates the combined use of the above-mentioned ginseng genus medicinal materials to prepare an active holographic ginsenoside composition (active holographic ginsenoside combination, AHGC) comprising mainly active panaxatriol saponins and glycol saponins. However, the preparation of such an active composition (AHGC) first requires the definition of the content ratio between panaxatriol saponin and panaxadiol saponin; second, it must be understood that central hyperexcitability and/or insufficient inhibitory function are closely related to the expression of clinical symptoms of Parkinson's Disease (PD) and the progression of the disease, as well as to the occurrence and progression of other neurodegenerative diseases including alzheimer's disease (AD, senile dementia) and amyotrophic lateral sclerosis (ALS, progressive freezing disease), etc. The action of panaxatriol saponin in promoting central excitability is unfavorable for the holographic ginsenoside composition to exert its efficacy in preventing and treating PD and other neurodegenerative diseases, and the action of panaxatriol saponin in resisting the composition in promoting central inhibitory action is also possible, so that the efficacy of the composition is weakened. However, both panaxatriol saponins and panaxadiol saponins have been reported to have antioxidant, anti-inflammatory and neuroprotective pharmacological effects, suggesting that the combined use of these two saponins may exert their own better efficacy alone. It can be seen that finding a proper ratio when the two kinds of saponin components are combined is important to whether the holographic ginsenoside composition integrates the pharmacological actions of the two components so as to improve the respective medicinal value. In order to find/reveal the appropriate ratio of the two contents in the composition, studies of example 2 and example 3 were conducted.
Example 2. Panaxadiol saponin composition and triol saponin composition were prepared in a ratio of 1 part of total saponins of American ginseng root and 2 parts of total saponins of American ginseng stem and leaf.
According to the measurement results of example 1, the commercial American ginseng root total saponins and American ginseng stem and leaf total saponins in 2020 were first comprehensively utilized and fed in a weight ratio (1:2), and the ginseng diol saponin composition (Panaxadiol saponins combination, PDSC 1) and the ginseng triol saponin composition (Panaxatriol saponins combination, PTSC 1) were prepared by separation by a conventional method. HPLC measurement data shows that PDSC1 contains five ginsenosides GRb, GRc, GRb2, GRb, and GRd, the total content of these 5 components is 93.03%, while PTSC1 mainly contains panaxatriol saponins GRg1 and GRe, the total content of these two saponins is 91.01%. The content of each individual ginsenoside is shown in Table 3, and HPLC analysis chromatograms of PDSC1 and PTSC1 are shown in FIGS. 6 and 7, respectively.
Table 3 content of main individual components of PDSC1 and PTSC1 (%, n=3)
Figure BDA0004007353830000172
Example 3. Discovery of rules of compatibility of Active Holographic Ginsenoside Compositions (AHGC) with pharmacological actions.
Parkinson's Disease (PD) is the second most common neurodegenerative disease that is secondary to alzheimer's disease (AD, senile dementia) and has a more mature animal model than other neurodegenerative diseases. Therefore, two internationally recognized models were first used to examine the efficacy of PDSC1 and PTSC1 and complexes of both at different dose ratios for treating Parkinson's Disease (PD) to determine their effects on efficacy from differences in efficacy and interactions. These two models are the model of mouse stiffness symptom induced by the anti-schizophrenia drug Haloperidol (HAL) through inhibition of dopamine receptor and the model of rat Parkinson's disease induced by Rotenone (ROT) through inhibition of mitochondrial complex enzyme I, which reflect the palliative and palliative effects of the test drug on PD, respectively. The former can simulate the movement symptoms caused by insufficient dopamine signals of PD patients, is commonly used for evaluating the efficacy of the drug to be tested for symptomatic treatment, and can respond to the non-dopamine mechanism of the efficacy, so that positive efficacy indicates that the drug to be tested can control or slow down the movement symptoms of PD, particularly the movement symptoms of advanced PD and atypical PD which are insensitive to dopamine drugs, and can also indicate that the drug to be tested can control or relieve the efficacy of PD-like movement symptoms, namely therapeutic PD syndrome, caused by drug treatment, particularly anti-schizophrenia drug treatment. The ROT model highly simulates the occurrence and development of PD and clinical characteristics from pathogenic factors, pathogenic mechanisms and symptoms, so that the ROT model is commonly used for evaluating the drug effect of the drug for preventing and treating the occurrence and development of PD, namely the effect of treating the root cause of PD, and researching the action mechanism of the drug.
Example 3-1.PTSC1 efficacy against or helping to slow down the symptoms of PD exercise depend on the relative proportions of the two.
The efficacy of the test agents was evaluated using the HAL-induced mouse stiffness model in the conventional manner. In summary, male ICR adult mice, which are adapted to the environment in the laboratory for 3-4 days and have a weight of 24+ -2 g, were subjected to pole climbing training once a day for 4 consecutive days to ensure that each animal school completed pole climbing tasks. The pole climbing test was completed on day 5 to obtain the evaluation stiffness parameters before molding, i.e., the time for the mouse head to rotate from top to bottom (Tturn) at the top of the pole and the time to climb completely from the top of the pole to the ground (Ttotal), calculated in seconds(s), allowing the pole climbing test to be completed within 90s, and if the above actions were not completed within 90s, then taken as 90s data. Then, the test agent was administered by gavage and HAL (2 mg/kg) was injected intraperitoneally 45 minutes later, and a stick test was performed between 45 minutes and 90 minutes after HAL administration to obtain experimental parameters of the model group and the test agent group.
Experimental results (table 4) show that PDSC1 induced the appearance of stiff symptoms against HAL (2 mg/kg) in a dose-dependent manner, in particular, the low dose (40 mg/kg) showed a trend of drug efficacy (p < 0.05), whereas both the medium dose (60 mg/kg) and the high dose (80 mg/kg) had very significant drug efficacy (p < 0.001).
Table 4 effect of panaxadiol saponin composition (PDSC 1) on anti-Haloperidol (HAL) induced stiffness (mean±sem, n=8)
Figure BDA0004007353830000181
Note that experimental data were analyzed by a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, &&& p<0.001; compared to HAL group after HAL administration, p<0.05,***p<0.001。
However, the PTSC1 composition was unable to combat H at doses of both 30mg/kg and 60mg/kgAL caused stiff symptoms, but all showed a trend to exacerbate stiff symptoms (table 5). On this basis, it was further observed that animals received both an effective dose of PDSC1 and different doses of PTSC1 against HAL induced stiffness, to determine whether PTSC1 impaired the efficacy of PDSC1 to slow HAL induced stiffness, or that both could be mixed in a specific content profile. The mixed dosing regimen included (1) HAL (2 mg/kg) +PDSC1 (60 mg/kg, optimal effective dose) +PTSC1 (60 mg/kg), (2) HAL (2 mg/kg) +PDSC1 (60 mg/kg) +PTSC1 (20 mg/kg), (3) HAL (2 mg/kg) +PDSC1 (30 mg/kg) +PTSC1 (30 mg/kg) (60 mg/kg total saponins) to determine the efficacy of the two saponin compositions in each ineffective dose aliquot to achieve a total saponin dose equivalent to the optimal dose of PDSC1. As shown in Table 5, the low dose PTSC1 (20 mg/kg) was administered in combination with the optimum dose of PDSC1 (60 mg/kg), and PTSC1 was able to reduce the effect of PDSC1 # p<0.05 With medium dose of PTSC1 (30 mg/kg) and equal dose of PDSC1 (30 mg/kg), PTSC1 can significantly weaken the drug effect of PDSC1 ## p<0.01 While high dose PTSC1 (60 mg/kg) is mixed with optimum dose of PDSC1 (60 mg/kg) in equal dose, PTSC1 almost completely counteracts the drug effect of PDSC1 ## p<0.01)。
Accordingly, the effect of a series of low doses of PTSC1 on the efficacy of the optimum dose of PDSC1 (60 mg/kg) was further determined, including administration of 1/2, 1/3, 1/4, 1/5 and 1/6 doses of PTSC1 and the optimum dose of PDSC1 mixture at the optimum dose of PDSC1 (60 mg/kg) to determine whether or not the proportion of the panaxatriol saponin composition exerting the proper efficacy exists when the panaxadiol saponin is the dominant composition of the holographic saponin composition. The results of the experiment (Table 6) show that when administered as a PTSC1/PDSC1 (1/2) mixture, the efficacy against stiffness in mice was comparable to that of the optimum dose of PDSC1, indicating that inclusion of one third of the PTSC1 in the mixture did not affect the efficacy of PDSC1 against the symptoms of PD. However, administration of the PTSC1/PDSC1 (1/3) mixture, PTSC1 significantly impaired the drug effect of PDSC1 & p<0.05 Other lower proportions of PTSC1 do not significantly impair the efficacy of PDSC 1.
Table 5 effect of panaxatriol saponin composition (PTSC 1) on Haloperidol (HAL) induced stiffness (mean±sem, n=8)
Figure BDA0004007353830000182
Figure BDA0004007353830000191
And (3) injection: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, &&& p<0.001; compared with the HAL group after HAL administration, *** p<0.001; compared with the HAL+PDSC1 (2+60) group after HAL, # p<0.05, ## p<0.01。
table 6 effect of low dose panaxatriol saponin composition (PTSC 1) on the efficacy of optimum dose panaxadiol saponin composition (PDSC 1) against stiff symptoms (mean±sem, n=10)
Figure BDA0004007353830000192
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p is less than 0.001; compared with the HAL group after HAL administration, # p<0.05, ## p<0.01, ### p<0.001; in comparison with the HAL + PDSC1 group, & p<0.05.
finally, the efficacy of a series of mixtures of PDSC1 and PTSC1 (wherein PDSC1 is the predominant amount and the total dose of these two compositions is 60 mg/kg) was further determined to determine whether one or more holographic ginsenoside compositions formulated at specific levels could be used in place of PDSC1 to combat HAL-induced stiffness. The results of the study are shown in Table 7, where the efficacy of the mixture at doses of 10mg/kg and 12mg/kg of PTSC1 was weaker than that of the control group of PDSC1, the efficacy was further reduced as the content of PTSC1 in the mixture increased until it was completely ineffective.
From the results of the above examples, it is clear that the panaxadiol saponin composition (PDSC 1) can very significantly combat HAL-induced rigidity in mice and thus can be used as an effective pharmaceutical form of ginsenoside for alleviating motor symptoms of PD. The panaxatriol saponin composition (PTSC 1) is not able to combat HAL-induced rigidity in mice, but instead tends to exacerbate rigidity. An equivalent dose of PTSC1 may completely cancel the efficacy of PDSC1, and PTSC1 at a lower dose than PDSC1 may impair the efficacy of PDSC1, but there is an example in which PTSC1 does not interfere with the efficacy of PDSC1 when the ratio of PTSC1/PDSC1 is 1/2. It can be seen that the effect of the interaction of PTSC1 and PDSC1 or the ratio of the two contents on the efficacy of the holographic ginsenoside composition is not linear, and the discontinuous jump change breaks the conventional knowledge of the person skilled in the art. Importantly, the anti-routine discovery can not only guide people to scientifically use the efficacy of ginseng and ginseng genus to inspire that people further excavate the medicinal value of ginsenoside.
It is noted that the total saponins of American ginseng root and the total saponins of American ginseng stem and leaf are calculated according to the weight of 1:2 and the weight ratio of PDSC1 to PTSC1 obtained by the batch preparation was 2.18. Therefore, we consider that the holographic ginsenoside composition formed according to the natural proportion is also an effective medicinal form for treating the symptoms of PD, and this point is verified in the subsequent study.
Table 7 efficacy of holographic ginsenoside against Haloperidol (HAL) to induce stiffness in mice with PDSC as the dominant composition (mean±sem, n=10)
Figure BDA0004007353830000193
Figure BDA0004007353830000201
/>
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p is less than 0.001; compared with the HAL group after HAL administration, # p<0.05, ## p<0.01, ### p<0.001; in comparison with the HAL + PDSC1 group, & p<0.05, && p<0.01, &&& p<0.001。
example 3-2.PTSC1 efficacy against or helping to assist PDSC1 against the development and progression of PD depends on the relative proportions of the two.
The rat PD model was induced by back subcutaneous injection of Rotenone (ROT) and progressive increase of dose, and the therapeutic effect of the test agents was evaluated using weight loss, PD clinical signs score, forelimb ability and motor balance ability as indicators, and the degeneration of the dopamine neurotransmission in the nigrostriatal was further determined for representative test groups.
Modeling method and disease degree assessment: the injection of rotenone was started at a low dose, with 25% dose increments every 5 days, once a day in the morning and evening. The specific method comprises the following steps: the first 5 days of modeling dose was 0.5mg/kg, the second 5 days of dosing increased to 0.625mg/kg, the third 5 days of modeling dose was 0.75mg/kg, and the volumes of each dosing were 0.2mL/100g, each time early (8:30, am) and late (20:30, pm). In experiments, the injection of rotenone is stopped when the model animals show 4 or more behavioral symptoms, and if the rats still have animals which do not reach 4-level behavioral manifestations after the third 5 days, the model is continuously molded according to the administration dosage of the third 5 days. Most animals typically exhibit PD behaviors of grade 3 and above for 12-15 days.
Classification level of clinical behavioral sign manifestations of PD model: a total of 6 levels. Stage 1: the hair is refused to be caught, the hair color becomes yellow and dirty, the bow back, the behavior is weakened, and the main activities are reduced; 2 stages: the primary symptoms are presented, but the primary activities are obviously reduced, the action is slow, and the tremor gait is unstable; 3 stages: the walking machine has secondary manifestation symptoms, and the gait is unstable or can not walk in a straight line or rotates to one side during walking; 4 stages: paralysis of front limb or hind limb, difficulty in walking and eating; 5 stages: one-sided recumbent, limb contracture and weight loss are great; stage 6: dying or dying.
Forelimb lift test (RT): the timing was started after the rats were placed in a black opaque cylindrical barrel, and the number of lifts of the forelimbs of the rats was recorded and observed for 5 min. The basis of the lifting of the forelimbs of the rats is that the forelimbs on one side or two sides are lifted over the shoulders and are contacted with the barrel wall until the forelimbs are completely put down and contacted with the barrel bottom, and then the lifting action is calculated.
Stride test (FAS): one hand of the experimenter fixes and supports the bones of the hind limbs of the rat body, fixes the right forelimb, enables the left forelimb of the rat to land, and ensures that the whole body weight falls on the left forelimb when the body and the tabletop form an angle of about 45 degrees. The rat was moved forward at a constant speed along the table edge at a distance of 90cm in 10s, and the number of times the left forelimb was lifted by the rat during the movement for 10s to maintain the body balance was recorded. Each rat was repeatedly assayed 3 times at 5s intervals, and the experimental results were averaged over 3 assays.
Stick rotation test (Rotarod): the rotation speed of the rat fatigue bar rotating instrument is set to be 15rpm, and the time is set to be 2min. After the instrument is accelerated to a set rotation speed, the head of the rat is placed on the rotating rod in the direction opposite to the rotation direction of the rotating rod, and four limbs of the rat are attached to the rotating rod. The time of the movement of the rat on the rotating rod, which is displayed by the instrument automatically stopping timing, is recorded. Each rat was measured 3 times, and the average value was taken as a measurement value at intervals of 2 minutes. The forelimb lifting test and the stride test can reflect the movement functions of the forelimbs of the animals, and the stick rotating test can reflect the limb movement functions and the whole body movement coordination capacity of the animals.
Test grouping, dosing and efficacy observation: adult male SD rats weighing 280-320 g, were randomly divided into a normal group, a Rotenone (ROT) model group and a test drug treatment group after being acclimatized. The test agent was administered once daily (8:30 am) and once daily (20:30 pm) by gavage each at a dose of half the total daily dose, 1 hour prior to rotenone administration, each administration having a volume of 0.2mL/100g. Normal control and rotenone model control were given equal volumes of physiological saline. The body weight and clinical behavior physical signs are monitored every day, and the above 3 tests are performed according to the set time point to quantitatively analyze the limb movement and coordination balance ability so as to judge the drug effect of the tested drug against the rotenone induced PD formation. When the animal behavior scoring grade of the model group reaches 3 grade or above, rotenone is not injected any more, and after various behavior indexes are collected after the day and 48 hours, brain tissues are fixed through heart perfusion paraformaldehyde for analyzing the degeneration condition of the nigrostriatal dopamine nerve pathway. At the same time, a parallel treatment was performed on the equivalent number of animals in each drug treatment group, and the treated animals were started with the highest behavioral score.
Based on the results of the study of example 3-1, the first observed test agent included three dose groups of PDSC1 (20, 40 and 60 mg/kg) and three dose groups of PTSC1 at the corresponding doses. As shown in Table 8, the result of the study shows that PDSC1 has dose-dependent effect of resisting rotenone to induce rats to form PD, the dose-effect relationship is a common inverted U-shaped dose-effect relationship, the effect is shown to be the best effect on all measured indexes by medium dose (40 mg/kg), and the effective dose range is 40-60 mg/kg. However, the efficacy of PTSC1 at 40mg/kg alone was comparable to that of an equivalent dose of PDSC1, while the other doses were ineffective. It can be seen that the effective dose range of PDSC1 against rotenone-induced PD formation is wider than the effective dose range of PTSC 1.
Next, the efficacy of the best effective dose of both combinations and the best dose of PDSC1 in combination with 1/2 dose of PTSC1 was observed to verify the results of the study in the HAL-induced mice stiffness model. As shown in table 8, both doses were 40mg/kg with simultaneous administration without any apparent efficacy, demonstrating that equal doses of PTSC1 can significantly counteract the efficacy of PDSC 1; when 40mg/kg of PDSC1 was combined with 20mg/kg of PTSC1, PTSC1 did not significantly impair the potency of PDSC 1. It can be seen that the holographic ginsenoside composition obtained by mixing PDSC1 and PTSC in equal ratio has no efficacy against PD occurrence and development, but the efficacy intensity of the holographic ginsenoside composition obtained by mixing 2 parts of PDSC1 and 1 part of PTSC1 is equivalent to the efficacy intensity of the equal dose of PDSC 1. In accordance with the effect of treating the symptoms of PD, the research result shows that the holographic ginsenoside composition formed according to the natural proportion of the panaxadiol soap is also an effective medicinal form for treating the root cause of PD.
TABLE 8 Effect of PDSC and PTSC in combination on Rotenone (ROT) induced formation of PD in rats
Figure BDA0004007353830000211
Note that: experimental data were analyzed using a one-way analysis of variance method. In comparison with the normal group, *** p<0.001; in comparison with the set of ROT s, ## p<0.01, ### p<0.001; compared with the PDSC1 (40 mg/kg) group, & p<0.05。
the results of the above two models are combined to demonstrate that the Holographic Ginsenoside Composition (HGC) constructed according to the ratio of PDSC1 to PTSC1 of 2:1 has equivalent pharmacological activity to treat both manifestation and root cause of PD compared with the equivalent dose of PDSC1, and therefore is hereinafter referred to as Active Holographic Ginsenoside Composition (AHGC). From the viewpoint of traditional Chinese medicine properties, the panaxatriol saponin composition (PTSC) is the material basis of seven parts of yang of the ginseng properties, while the panaxadiol saponin composition (PDSC) is the material basis of three parts of yin thereof, so that AHGC is a yin-yang combination with the advantage of negative. For diseases, such as PD, from pathogenesis to disease symptom expression is closely related to hyperexcitability and insufficient inhibitory function, but the development of excitatory acetylcholine signals is reduced with the prolongation of the course of the disease, and long-term administration is required. Therefore, the AHGC is a scientific, safe and effective medicinal mode from the viewpoint of traditional Chinese medicine theory and modern medicine. In addition, AHGC is used as a medicinal mode, the dosage of the panaxadiol saponins is reduced by about 1/3, so that the resource consumption of raw medicinal materials can be obviously reduced. Obviously, compared with the medicinal mode in which total saponins, glycol saponin compositions or single saponins are formed, AHGC is not only a brand new mode, but also a novel mode of medication in which rare medicinal material resources are consumed with low consumption and high drug effect are expressed. More importantly, the content ratio of PTSC/PDSC in the AHGC and the content configuration of main active ingredients GRg, GRe, GRb1, GRc, gb2, GRb3 and GRd in the PTSC/PDSC content ratio provide a feeding basis and a product quality standard for preparing the active holographic ginsenoside composition by comprehensively utilizing ginseng, american ginseng and pseudo-ginseng.
Examples 3-3. Determination of the content profile of the major individual components of Active Holographic Ginsenoside Composition (AHGC) to define the content profile of the major active individual components of the preferred composition.
Accordingly, the content of total saponins, panaxatriol saponins and panaxadiol saponins in holographic ginsenoside compositions (AHGC) prepared by different proportions of PDSC1 and PTSC1 and the content of the 7 saponins were determined by HPLC analysis, and the relationship between the content configuration and the efficacy was intuitively analyzed by combining the efficacy of the HAL and ROT models. The experimental results are shown in table 9, and these experimental data will be used as guiding index for the subsequent preparation of AHGC.
TABLE 9 Active Holographic Ginsenoside Composition (AHGC) Main Single ingredient content compounding and pharmacodynamic relationship
Figure BDA0004007353830000212
Figure BDA0004007353830000221
Note that: TPDS, total panaxadiol saponins; TPTS: total panaxatriol saponins; + corresponds to p<0.05, ++p<0.01 of the total number of the components, ++ pair should p<0.001; a The dosage range is narrow.
Example 4 comprehensive utilization of ginseng, american ginseng and notoginseng as ginseng medicinal materials to prepare Active Holographic Ginsenoside Composition (AHGC)
According to the above-mentioned guiding parameters of AHGC content configuration and content data of 7 ginsenosides among total saponins of ginseng root and stem and leaf, total saponins of American ginseng root and stem and leaf, and total saponins of notoginseng stem and leaf obtained in example 1, the following 10 dosing schemes were designed to prepare 10 samples of holographic ginsenoside compositions (HGC 1 to HGC 10): (1) preparing HGC1 from a mixture of 1 part of American ginseng root total saponin and 2 parts of American ginseng stem and leaf total saponin, (2) preparing HGC2 from a mixture of 1 part of American ginseng root total saponin and 1 part of American ginseng stem and leaf total saponin and 3 parts of American ginseng stem and leaf total saponin, (3) preparing HGC3 from a mixture of 2 parts of ginseng root total saponin and 1 part of ginseng stem and leaf total saponin and 1 part of pseudo-ginseng stem and leaf total saponin, (4) preparing HGC4 from a mixture of 2 parts of ginseng root total saponin and 1 part of ginseng stem and leaf total saponin and 2 parts of pseudo-ginseng stem and leaf total saponin, (5) preparing HGC5 from a mixture of 1 part of ginseng root total saponin, 1 part of ginseng stem and leaf total saponin and 1 part of notoginseng stem and leaf total saponin, (6) preparing HGC6 from a mixture of 1 part of ginseng root total saponin, 1 part of ginseng stem and leaf total saponin and 2 parts of notoginseng stem and leaf total saponin, (7) preparing HGC7 from a mixture of 2 parts of American ginseng root total saponin, 1 part of American ginseng stem and leaf total saponin and 1 part of notoginseng stem and leaf total saponin, (8) preparing HGC8 from a mixture of 2 parts of American ginseng root total saponin, 1 part of American ginseng stem and leaf total saponin and 2 parts of notoginseng stem and leaf total saponin, (9) HGC9 is prepared from a mixture of 1 part of American ginseng root total saponin, 1 part of American ginseng stem and leaf total saponin and 1 part of pseudo-ginseng stem and leaf total saponin, and HGC10 is prepared from a mixture of 1 part of American ginseng root total saponin, 1 part of American ginseng stem and leaf total saponin and 2 parts of pseudo-ginseng stem and leaf total saponin.
The preparation method comprises the following steps: 30 g of the mixture according to the dosing scheme are dissolved in 300 ml of 30% ethanol aqueous solution, respectively, and are applied to a reversed phase C equilibrated with 30% ethanol 18 Silica gel (ODS, 300 g) column. The elution was performed with 2.0L of 30% aqueous ethanol, one fraction was collected per 500 ml, and a total of 4 fractions (Fr.1-Fr.4) were collected, and then with 5.0L of 55% aqueous ethanol, one fraction was collected per 500 ml, and a total of 10 fractions (Fr.5-Fr.14) were collected. The components were tested by HPLC and the components (Fr.4-Fr.12) containing ginsenosides GRg1, GRe, GRb1, GRc, GRb2, GRb and GRd were combined and concentrated under reduced pressure to complete dryness to give holographic ginsenosides HGC1 (24.60 g, 82.0% yield), HGC2 (23.66 g, 78.9% yield), HGC3 (23.16, 77.2% yield), HGC4 (22.70, 75.7% yield), HGC5 (23.48, 78.3% yield), HGC6 (22.80, 76.0% yield), HGC7 (24.56, 81.9% yield), HGC8 (23.61, 78.7% yield), HGC9 (24.63, 82.1% yield) and HGC10 (22.9) yield 76.3% respectively. The content of the 7 ginsenosides in these components was then determined by HPLC method, and their relative proportions were analyzed.
Table 10 ten feeding schemes comprehensive utilization of total saponins of ginseng, american ginseng and notoginseng to obtain holographic ginsenoside compositions (HGC 1 to HGC 10) with content of each saponin (%, n=3)
Figure BDA0004007353830000222
Figure BDA0004007353830000231
The results of the study are shown in Table 10 and FIGS. 8-17, and 10 holographic ginsenoside compositions (HGC 1-HGC 10) prepared by 10 different feeding schemes all meet the following conditions: the total amount of the panaxatriol saponins and the panaxadiol saponins is more than 80 percent, wherein the total amount of the panaxadiol saponins is 1.8 to 4.4 times that of the panaxatriol saponins; the contents of each of five ginsenosides GRe, GRb1, GRc, GRb3 and GRd are more than 5% and less than 25%, but the case that the contents of the five ginsenosides are simultaneously more than 15% is excluded; GRg1 is present in an amount of between 2.80 and 7.71% and GRb2 is present in an amount of between 4.72 and 10.45%.
Next, the efficacy of the above 10 ginsenoside compositions was examined using HAL-induced mice stiffness model that can respond to the efficacy of the drug against motor symptoms (i.e., palliative) and Rotenone (ROT) -induced rat PD model that reflects the efficacy of the drug against PD occurrence and development (i.e., palliative). As shown in Table 11, the administration dosage of 60mg/kg of the 10 ginsenoside compositions can obviously resist the symptoms of the HAL induced mice to appear stiff ### p<0.001 And the potency of each composition did not differ significantly.
Table 11 influence of ten dosing regimens on HAL-induced mouse stiffness by Holographic Ginsenoside Composition (HGC) (mean±sem, n=10)
Figure BDA0004007353830000232
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p<0.001; compared with the HAL group after HAL administration, ### p<0.001。
however, in the model of ROT-induced rat PD, a significant difference in potency between these 10 compositions occurred at the dose of 60mg/kg administered (table 12). Of these 10 compositions, three of HGC6, HGC7 and HGC8 had no significant protective efficacy, while the other 7 effective compositions had different protective efficacy strengths, with the 4 compositions HGC1, HGC2, HGC4 and HGC9 having comparable efficacy strengths, almost all of which were able to completely combat ROT-induced formation of PD in rats, HGC3 and HGC5 having inferior efficacy strengths, and HGC10 having inferior efficacy strengths.
Results of research on two animal models of HAL and ROT prove that HGC1 to HGC5, HGC9 and HGC10 are effective compositions for treating both symptoms and root causes of PD, wherein HGC1, HGC2, HGC4 and HGC9 are more prominent; in combination with the content configuration of each ginsenoside component in each composition (Table 10), these efficacy results again demonstrate that any adducts of each active ginsenoside of ginsenosides GRg, GRe, GRb1, GRc, GRb2, GRb3 and GRd have no efficacy guarantee for the prevention and treatment of PD, and that proper content ratios therebetween may be a key issue in determining efficacy intensity. This possibility will be verified in subsequent studies.
Table 12 influence of ten feeding schemes on Rotenone (ROT) -induced rat PD formation by Holographic Ginsenoside Composition (HGC) (mean±sem, n=7)
Figure BDA0004007353830000241
Note that: experimental data were analyzed using a one-way analysis of variance method. In comparison with the normal group, *** p<0.001; in comparison with the set of ROT s, # p<0.05, ## p<0.01, ### p<0.001。
the results of example 4 are summarized: for a long time, anti-PD drugs are symptomatic treatments in the global scope, and no drugs capable of preventing and treating the occurrence and development of PD exist up to now. Furthermore, dopamine-based drugs are ineffective in freezing gait and posture imbalance (easy fall) in patients with advanced PD. It can be seen that those holographic ginsenoside compositions (AHGC) having significant efficacy on both HAL-induced stiffness models (simulating gait freezing and posture imbalance in patients with middle and late stage PD) and ROT-induced PD occurrence and development models, including HGC1 to HGC5, HGC9 and HGC10 (particularly HGC1, HGC2, HGC4 and HGC 9), can exert both principal and secondary aspect of the PD effect, and have significant pharmaceutical value for controlling PD, and therefore, these compositions are referred to as "optimal compositions" and those subsequently discovered to have significant efficacy on both of the above-mentioned two PD animal models are collectively referred to as "optimal compositions". Therefore, the above-described feeding schemes (1), (2), (3), (4), (5), (9) and (10) can be used as preferred schemes for preparing the optimum composition, with (1), (2), (4) and (9) being preferred. Of course, in order to avoid the difference of the content of the effective components caused by the raw materials in different years, the preferable feeding scheme can be correspondingly adjusted according to the actual situation.
In particular, the research results indicate that the proper content ratio of the different active ginsenosides in the composition is a key problem for determining the strength of the medicament effect. Thus, it is further disclosed that this suitable ratio is critical to the preparation of a quality stable, optimal composition. Therefore, the difference of the effective pharmaceutical strengths of the different holographic ginsenoside compositions can be exactly used for analyzing the relationship between the pharmaceutical efficacy and the content configuration of each main ginsenoside component in the composition so as to reveal the content configuration rule of the effective components of the effective composition.
Example 5. Analysis of the relationship between the content of each main ginsenoside component in Holographic Ginsenoside Composition (HGC) and the efficacy.
Example 5-1. The content of panaxadiol saponin Rb2 (GRb 2) in HGC did not affect the efficacy of HGC.
As shown in table 10, the total saponin content of each composition and the total ginsenoside content of the panaxadiol saponin were all at the same level, excluding the effect of these two factors on the efficacy of different holographic ginsenoside compositions, thus suggesting that the content configuration of different effective individual components is critical. Further, the study data (tables 10 and 12) suggest that the content of panaxadiol saponins (GRb 2) does not greatly contribute to the efficacy of the composition against HAL-induced stiffness symptoms and ROT-induced PD formation, and thus the possibility of being one of the main active ingredients of the efficacy can be primarily excluded.
To verify this possibility, the applicant investigated the efficacy of PDSC1 (PDSC 1-GRb 2) lacking GRb at the optimal dose (60 mg/kg in HAL model and 40mg/kg in ROT model) by contrast using the feature that PDSC1 is effective in both palliative (HAL-induced mouse stiffness model) and palliative (ROT-induced rat PD model) models. According to the main constituent content of PDSC1, GRb2 was contained in an amount of 4.73mg per kg of the dose, and thus the dose of the group PDSC1-GRb2 was 55.27mg per kg. Similarly, the corresponding dose of PDSC1-GRb2 was 36.85mg/kg in the 40mg/kg dose of PDSC 1.
The results of the studies are shown in tables 13 and 14, and the efficacy of the PDSC1 and the PDSC1-GRb are not significantly different for the models for treating both the symptoms and root causes, which further proves that GRb is not a main active ingredient of PDSC1 for treating both the symptoms and root causes of PD, and the primary content thereof does not affect the efficacy of the composition. Therefore, in preparing the optimum composition, the GRb content may allow the optimum composition to appear in a raw state for simplifying the process flow, and GRb components may not be considered in the subsequent study of the compatibility rules among the ginsenoside active components in the optimum composition.
Table 13 efficacy of PDSC1 deleted for GRb in the pole-climbing assay against Haloperidol (HAL) to induce stiffness in mice (mean±sem, n=10)
Figure BDA0004007353830000251
And (3) injection: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p<0.001; compared with the HAL group after HAL administration, ### p<0.001。
table 14GRb effects of deleted PDSC1 on Rotenone (ROT) -induced rat PD formation (mean±sem, n=5
Figure BDA0004007353830000252
Note that: experimental data were analyzed using a one-way analysis of variance method. In comparison with the normal group, *** p<0.001; in comparison with the set of ROT s, ### p<0.001。
example 5-2. Analysis of the relationship between the content configuration of the main individual components and the efficacy of the Holographic Ginsenoside Composition (HGC).
Based on the above results, the relationship between the relative content configuration of two kinds of panaxatriol saponins (GRg and GRe) and four kinds of panaxadiol saponins (GRb, GRc, GRb3 and GRd) and the efficacy of the composition was visually analyzed using the study data list of example 4 (table 15). The content of GRe in the composition is far higher than GRg1, and the biological activity and pharmacological action intensity of the two single components are known to be similar, so GRe is representative of panaxatriol saponin in the composition, and GRe is taken as a functional unit when the content configuration and the pharmacodynamic relation of the main single components in the composition are analyzed; GRb1 is known to be converted to GRd in the gut by the gut flora, thus classifying GRb1 and GRd as the same functional unit and GRc and GRb as another functional unit.
Accordingly, the applicant set ratios (hereinafter abbreviated as 8 ratios) of TPDS/TPTS (panaxadiol total saponin/panaxadiol total saponin), GRe/GRg1, GRb/GRe, GRb1/GRd, GRb1/GRc, GRb1/GRb3, GRc/GRb3 and (GRb 1+ GRd)/(GRc + GRb 3) to reveal the relationship between the content configuration of the main individual components and the efficacy of the Holographic Ginsenoside Composition (HGC). Judging the effective level according to the statistical result of the experimental data, wherein p is more than 0.05 and is ineffective; p <0.05, active (+); p <0.01, very potent (++); p is less than 0.001, and the total number of the samples is less than 0.001, very much there is effect (+ ++). The efficacy intensity of the ROT model is based on the efficacy of the last dose of ROT on the next day after the last dose.
As shown in table 15, for each of the holographic ginsenoside compositions (HGC 1 to HGC 10) that were very effective against HAL-induced PD stiffness, the eight ratios were allowed to be: TPDS/tpts=1.88-4.40, GRe/GRg 1=2.31-4.41, GRb 1/gre=0.64-1.86, GRb1/GRd =0.79-2.08, GRb1/GRc =0.67-2.17, GRb1/GRb 3=0.82-2.76, GRc/GRb 3=0.79-2.11, (GRb 1+ GRd)/(GRc + GRb 3) =0.66-1.92. Notably, for each of the holographic ginsenoside compositions (HGC 1-HGC 5, HGC9 and HGC 10) which were very effective against ROT-induced rat PD formation, the range of the other 7 ratios was narrowed in addition to the ratio GRc/GRb, respectively: TPDS/tpts=1.88 to 4.29, GRe/GRg1 =2.33 to 4.03, GRb 1/gre=0.64 to 1.53, GRb1/GRd =0.79 to 1.56, GRb1/GRc =0.75 to 2.17, GRb1/GRb 3=0.93 to 2.20, (GRb 1+ GRd)/(GRc + GRb 3) =0.76 to 1.92. Therefore, these 8 ratio ranges with excellent effect on the ROT model can be used as quality standards for preparing the effective Active Holographic Ginsenoside Composition (AHGC). HGC 1-HGC 5, HGC9 and HGC10 meet this quality standard, and their GRg/GRe/GRb 1/GRc/GRb3/GRd ratios are 0.60/2.13/2.17/1.00/1.27/2.12, 0.76/2.16/1.38/1.00/0.63/1.74, 0.37/0.86/1.00/1.00/0.47/0.64, 0.28/0.66/0.76/1.00/0.68/0.52, 0.39/0.91/0.75/1.00/0.56/0.66, 0.23/0.91/1.39/1.00/1.09/0.91 and 0.21/0.76/1.01/1.00/1.09/0.73, respectively. It can be seen that the relative proportions of the 6 ginsenoside active ingredients in these optimal compositions did not change regularly, and that these ratio ranges were not regionally different from the ratio ranges of GRg/GRe/GRb/GRc/GRb 3/GRd in those compositions (HGC 6-HGC 8) that had poor efficacy in the ROT model. Therefore, the quality standard of the effective composition cannot be formulated by using only the relative values of the contents of the 6 ginsenoside components, but the 8 ratio is required as the quality standard.
TABLE 15 Effect of the configuration of the content of the Main individual ginsenosides in the holographic ginsenoside composition on the efficacy
Figure BDA0004007353830000261
Figure BDA0004007353830000271
Note that: TPDS: total content of panaxadiol saponins; TPTS: total content of panaxatriol saponins. Judging the effective level according to the statistical result of the research data in the embodiment 4, wherein p is more than 0.05 and is ineffective; p <0.05, active (+); p <0.01, very potent (++); p is less than 0.001, and the total number of the samples is less than 0.001, very much there is effect (+ ++).
Conclusion(s)
The above parameters further clearly demonstrate the unpredictability of the 8 ratio in the preferred compositions, and each preferred composition has its own 8 ratio, which further highlights the anti-routine nature of the invention and its significant use value. Further, under the condition of certain content proportion, different active ginsenosides cannot cooperate with each other to generate obvious pesticide effect and even weaken respective pesticide effects mutually, so that a mixture of a plurality of single active ginsenosides is not guaranteed in pesticide effect. Similarly, the efficacy of the optimal composition is not the sum of the independent efficacy of the effective single components, but the result of the synergistic effect of the compatibility of different single components, and a reasonable 8-ratio is the essence of optimal compatibility. Obviously, these findings break the routine knowledge of those skilled in the art of utilizing the biological activity and pharmacological effects of ginsenoside, thereby advantageously avoiding ineffective and inefficient modes of ginsenoside use. More importantly, these findings reveal that the biological activity and pharmacological action of ginsenoside can be improved and expanded by the compatible use of active ginsenoside components, and the active ginsenoside composition has great potential for health care and disease treatment of ginseng (ginseng, american ginseng and pseudo-ginseng) and the ginsenoside contained in the ginseng, so that the optimal composition comprising HGC 1-HGC 5, HGC9 and HGC10 is not only an optimal compatibility for preventing and treating PD, but also can be applied to nervous system, cardiovascular system, immune system and endocrine system and systemic sub-health state thereof.
Example 6. Panaxadiol saponin composition contained in the excellent holographic ginsenoside composition has the effect of treating both principal and secondary aspect of PD.
In order to further develop the compatibility rules of the main active ginsenoside components which may exist in the effective holographic ginsenoside composition and show the drug effect, the applicant selects and researches the optimal compatibility or compatibility rules of four main active ginsenoside (GRb, GRc, GRb3 and GRd). To this end, first, the same dosing regimen for preparing the Holographic Ginsenoside Composition (HGC) was selected to prepare the corresponding panaxadiol saponin composition (PDSC) enriched only in panaxadiol saponins; these panaxadiol saponin compositions (PDSC) were then evaluated for their efficacy in treating PD using the two animal models previously used for both palliative and palliative measures.
Example 6-1. Preparation of panaxadiol saponin composition (PDSC) corresponding to Holographic Ginsenoside Composition (HGC).
More specifically, (1) PDSC1 is prepared from a mixture of "1 part of ginsenoside 1 + 2 parts of ginsenoside, PDSC2 is prepared from a mixture of" 1 part of ginsenoside 1 + 3 parts of ginsenoside 1 + 2 parts of ginsenoside, PDSC4 is prepared from a mixture of "2 parts of ginsenoside 1 + 2 parts of ginsenoside, 4) PDSC5 is prepared from a mixture of" 1 part of ginsenoside 1 + 1 part of ginsenoside 1 parts of stem and leaf, (5) PDSC6 is prepared from a mixture of "1 part of ginsenoside 1 + 2 parts of ginsenoside 2 parts of stem and leaf, (6) PDSC7 is prepared from a mixture of" 1 part of ginsenoside 1 + 2 parts of ginsenoside 1 "and 7), and (9) PDSC1 is prepared from a mixture of" 1 part of saponin 1 part of ginsenoside 1 + 2 parts of stem and leaf of ginsenoside "and 10 parts of PDSC 1. Wherein the holographic ginsenoside compositions HGC1, HGC2, HGC4, HGC5, HGC9 and HGC10 prepared by the feeding schemes (1), (2), (4), (5), (9) and (10) are excellent compositions, and the holographic ginsenoside compositions HGC6 and HGC7 prepared by the feeding schemes (6) and (7) have poor drug efficacy.
The preparation method comprises the following steps: 30 g of the mixture according to the dosing scheme are dissolved in 300 ml of 30% ethanol aqueous solution, respectively, and are applied to a reversed phase C equilibrated with 30% ethanol 18 Silica gel (ODS, 300 g) column. The eluate was first eluted with 2.0 l of 30% aqueous ethanol, the 30% aqueous ethanol eluate was discarded, then eluted with 3.0 l of 43% aqueous ethanol, and the eluate was collected in one portion per 500 ml, and 6 components (fr.1 to fr.6) were collected in total, and then eluted with 6.0 l of 55% aqueous ethanol, and 10 components (fr.7 to fr.18) were collected in total, per 500 ml. The components were examined by HPLC, and the components (Fr.7 to Fr.15) containing ginsenoside GRb1, GRc, GRb2, GRb and GRd were combined and concentrated under reduced pressure to complete dryness to give ginsenoside composition PDSC1 (15.16 g, yield 50.5%), PDSC2 (13.88 g, yield 46.3%), PDSC4 (16.77, yield 55.9%), PDSC5 (15.09, yield 50.3%), PDSC6 (16.12, yield 53.7%), PDSC7 (16.41, yield 54.7%), PDSC9 (17.01, yield 56.7%) and PDSC10 (16.68), yield 55.6%) respectively.
Table 16 content and total content of each saponin (%, n=3) in panaxadiol saponin composition (PDSC) corresponding to Holographic Ginsenoside Composition (HGC)
Figure BDA0004007353830000272
Figure BDA0004007353830000281
TABLE 17 arrangement of the contents of the respective major individual components in the panaxadiol saponin composition (PDSC) corresponding to the Holographic Ginsenoside Composition (HGC)
Figure BDA0004007353830000282
The contents of 5 kinds of panaxadiol saponin components of GRb, GRc, GRb2, GRb3 and GRd and their total contents in the panaxadiol saponin composition (PDSC) were measured by a conventional HPLC method, and the ratios of GRb1/GRd, GRb1/GRc, GRb1/GRb3, GRc/GRb3 and (GRb 1+ GRd)/(GRc + GRb 3) were calculated. Analytical measurements revealed that the total amount of 5 ginsenosides GRb1, GRc, GRb2, GRb and GRd in these 8 compositions constituted 91% or more of the main composition (Table 16 and FIGS. 18 to 25), and that the ratios GRb1/GRd, GRb1/GRc, GRb1/GRb3, GRc/GRb3 and (GRb 1+ GRd)/(GRc + GRb 3) in each composition (Table 17) were consistent with the ratios in the corresponding Holographic Ginsenoside Compositions (HGC) (Table 15), respectively.
Example 6-2. Efficacy of panaxadiol saponin composition (PDSC) corresponding to Holographic Ginsenoside Composition (HGC) on PD was measured.
The efficacy of PDSC1, PDSC2, PDSC4, PDSC5, PDFSF6, PDSC7, PDSC9 and PDSC10 against motor symptoms of PD caused by dopamine signal deficiency was determined using HAL-induced mouse stiffness model, while the efficacy of anticholinergic for motor symptom relief was compared and observed.
Table 18 Holographic Ginsenoside Composition (HGC) effect of corresponding panaxadiol saponin composition (PDSC) against HAL-induced stiffness in mice (mean±sem, n=10)
Figure BDA0004007353830000283
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, ** p<0.01, *** p<0.001; compared with the HAL group after HAL administration, ### p<0.001; in comparison with the PDSC1 group, & p<0.05。
study data (table 18) shows: these compositions were all very significant against the symptoms of HAL-induced stiffness in mice, but the potency of the different compositions differed, with PDSC1 and PDSC2 being the strongest and being both stronger than that of ambrisen, and the potency of each composition did not differ significantly from one another, and the potency (table 18) was consistent with that of the respective corresponding Holographic Ginsenoside Composition (HGC) (table 11). However, except for PDSC2, the potency was better than PDSC4, PDSC5, PDSC9 and PDSC910, while HGC2 was the opposite potency with respect to them, indicating that the panaxatriol saponin composition (PTSC) in HGC2 may impair the potency of the panaxadiol saponin composition (PDSC). Notably, the ratio of PDSC to PTSC in HGC3 was 3.06 (table 15), whereas just this ratio of PTSC1 mixed with PDSC1 constituted HGC having less potency than PDSC1, but the addition of PTSC1 at ratios greater or less than this determined did not significantly impair the potency of PDSC1 (table 6). This further indicates that the inclusion of 1/4 of PTSC in HGC is disadvantageous in alleviating motor symptoms caused by insufficient dopamine signals or elevated acetylcholine signals.
Taken together, the following conclusions can be drawn:
(1) The panaxadiol saponin composition (PDSC) is the main active ingredient of the Holographic Ginsenoside Composition (HGC) for resisting the symptoms of the rigidity induced by HAL to mice, and can be used as an ideal medicinal form for controlling the movement symptoms of PD.
(2) The potency of PDSC obtained by different feed ratios of the same medicinal materials is different, and the main content configuration of panaxadiol saponins is related to the potency of PDSC for relieving PD exercise symptoms, and the relationship between the two is revealed in subsequent researches.
(3) In most cases, the drug effect intensity of HGC and corresponding PDSC for alleviating PD movement symptoms is equivalent, and again, it is explained that HGC is also an effective drug form for alleviating PD movement symptoms, and 1/3 to 1/5 of the ginsenoside resource can be saved by taking HGC as a drug form.
(4) Since acetylcholine signal hyperactivity is the neurochemical substance of the symptoms of HAL-induced stiffness, the effect of PTSC in attenuating PDSC against HAL-induced stiffness reflects its pharmacological effect in promoting acetylcholine signal, whereas the neurodegenerative related deficiency in acetylcholine signal is closely related to cognitive impairment in advanced PD patients and also involved in cognitive impairment in senile dementia, which predicts that HGCs are more suitable for advanced PD patients and AD patients than PDSCs with weaker efficacy against HAL-induced stiffness, such as PDSC5 and PDSC 10. The corresponding total ginsenoside composition is also more suitable for improving hypomnesis of the normal elderly.
It is also pointed out that the medicinal effect of the anti-haloperidol (anti-schizophrenia) on inducing the mice to develop stiff symptoms also supports the medicinal use of the panaxadiol saponin composition for preventing and treating parkinsonism-like symptoms (DIP) caused by the drug treatment. DIP, also known as drug parkinsonism, is the second most common cause of parkinsonism following PD. Drug-induced DIP secondary to antipsychotics and other dopamine antagonists is common clinically and is insensitive to dopamine-like drugs, and DIP is currently slowed down clinically by discontinuation or use of anticholinergic agents with serious side effects. It can be seen that Holographic Ginsenoside Compositions (HGC), in particular panaxadiol saponin compositions PDSC1 and PDSC2, will provide a safer and more effective new way to control DIP than ambrisen.
The efficacy of those good acting HGCs and their corresponding PDSCs in the ROT model, including HGC1, PDSC1, HGC2, PDSC2, HGC4, PDSC4, HGC9 and PDSC9, was further determined using the ROT-induced rat PD model, while the efficacy of the two panaxadiol saponin compositions, namely, PDSC6 and PDSC7, corresponding to HGC6 and HGC7 (not good acting compositions) was studied in comparison. The dosage of each tested composition was 40mg/kg, as before.
As shown in table 19, other panaxadiol saponin compositions except PDSC6 and PDSC7 were very significantly resistant to formation of a PD model in the ROT-induced rats, and no significant difference was seen between the compositions, showing that all the indexes measured at two time points of the last day of administration of the last test drug and the third day of drug withdrawal, 2 days after drug withdrawal, were very significantly superior to the ROT control group, and were close to the normal control group. In particular, these superior PDSCs exhibited the same potency as their corresponding Holographic Ginsenoside Compositions (HGCs) (table 12). Consistent with HGC6 and HGC7, PDSC6 and PDSC7 only partially improve the locomotor behavior and precondition elevation capability on the rotating stick. The results of the study demonstrate that PDSC is an indispensable active ingredient of HAF against the formation of rat models of ROT-induced PD, and the content configuration of GRb, GRc, GRb3 and GRd is a key factor determining whether HGC and PDSC can resist the development of mitochondrial complex enzyme I deletion-induced PD and the efficacy intensity thereof.
Table 19 Holographic Ginsenoside Composition (HGC) and corresponding panaxadiol saponin composition (PDSC) against the effect of Rotenone (ROT) on induced rat PD formation (mean±sem, n=5)
Figure BDA0004007353830000291
Figure BDA0004007353830000301
Note that: experimental data were analyzed using a one-way analysis of variance method. In comparison with the normal group, *** p<0.001; in comparison with the set of ROT s, ### p<0.001; in comparison with the PDSC1 group, & p<0.05, && p<0.01。
examples 6-3 analysis of the content configuration and pharmacodynamic relationship of panaxadiol saponins GRb, GRc, GRb3 and GRd.
Here, the content configuration and the pharmacodynamic relationship of GRb, GRc, GRb3 and GRd are further visually analyzed by using the study data of example 6-1 and example 6-2, and the study results can provide quality standards for preparing quality-stable and controllable optimal compositions. As shown in Table 20, the purity of ginsenosides of different HGCs and corresponding PDSCs was varied between 4-10%, but this difference was not related to the poor efficacy of HGC6, PDSC6, HGC7 and PDSC7 in the ROT model, and the relative content between the ginsenosides was closely related to the efficacy intensity. The concrete steps are as follows: the ratio of GRb/GRd of the optimal composition (with very obvious drug effects on the HAL and ROT models) is between 0.79 and 1.53, the ratio of GRb1/GRc is between 0.76 and 2.17, the ratio of GRb1/GRb3 is between 1.11 and 2.20, the ratio of GRc/GRb3 is between 0.79 and 1.59, and the ratio of (GRb 1+ GRd)/(GRc + GRb 3) is between 0.76 and 1.92, and the optimal composition is within an optimal range. These analysis results are completely consistent with those of Table 15 (influence of the configuration of the content of the main individual ginsenosides in the holographic ginsenoside composition on the efficacy), and it can be further confirmed that the ratio of GRc/GRb3 is in the range of 0.79 to 1.59 by combining Table 15 and Table 20.
By combining the results of the studies of examples 3 to 6, the following 3 conclusions can be drawn:
(1) The panaxadiol saponins are indispensable effective components for treating both the symptoms and root causes of PD by using the holographic ginsenoside composition (HAF) and the panaxadiol saponin composition (PDSC).
(2) At the same dose, the efficacy intensity of the excellent HAF is equivalent to that of the corresponding PDSC for both the temporary and permanent cure, that is, the PDSC can exert the full dose of the excellent HAF and the excellent PDSC according to the HPLC measurement result (table 10) at the dose of 2/3 to 4/5 of the holographic ginsenoside composition (HAF). The holographic ginsenoside composition (HAF) is used as a medicinal form, and the dosage of the panaxadiol saponin composition (PDSC) can be reduced by 1/3 to 1/5, which has important value for relieving the shortage of ginseng, american ginseng and pseudo-ginseng medicinal materials, so that the active holographic ginsenoside composition (AHAF) is further supported to be an economic, efficient and scientific medicinal form. In particular, since the ability of the optimal panaxadiol saponin composition (PDSC) to correct central hyperexcitability is greater than that of the holographic ginsenoside composition (HAF) as analyzed from the drug properties, it is expected that PDSC will better serve those patients with prominent hyperexcitability than HGC, and that these patients should theoretically exhibit prominent PD motor symptoms. While the active ingredient GRe may confer a broader brain protective effect to HGC, HGC may be superior to PDSC in terms of efficacy against atypical parkinsonism. Therefore, HGC or PDSC can be flexibly used according to the disease and the patient's specific situation, but this needs to be verified by clinical trials.
(3) The content configuration of GRb, GRc, GRb3 and GRd is a key factor for determining whether HGC and PDSC can resist the development of PD induced by deletion of mitochondrial complex enzyme I and the efficacy intensity thereof. The 5 ratio of the content configuration of the 4 ginsenosides in the best-acting composition (very remarkable efficacy on both HAL and ROT PD models) has the following characteristics: GRb1/GRd ratio is between 0.79 and 1.53, GRb/GRc ratio is between 0.76 and 2.17, GRb1/GRb3 ratio is between 1.11 and 2.20, GRc/GRb3 ratio is between 0.79 and 1.59, and (GRb 1+ GRd)/(GRc + GRb 3) ratio is between 0.76 and 1.92.
TABLE 20 influence of the content configuration of the main panaxadiol saponins in HGC and its corresponding PDSC on the efficacy
Figure BDA0004007353830000311
Note that: study of data statistics according to example 6 to determine the level of validity, p >0.05, invalid; p <0.05, active/+; p <0.01, very effective/++; p is less than 0.001, and the total number of the samples is less than 0.001, very effective/++.
Example 7 effect of GRc/GRb3 ratio and (GRb 1+ GRd)/(GRc + GRb 3) ratio on the efficacy of PDSC.
On the basis of the above-mentioned optimum ranges, the applicant has sought to further find the better 5 ratios that may be present. For this purpose, taking PDSC1 as an example, high purity GRc and GRb3 were added to the best-effect PDSC1 to gradually change the 5 ratio, and changes in the efficacy of these new compositions were observed using HAL-induced mouse stiffness model and ROT-induced rat PD development model.
EXAMPLE 7-1 preparation of PDSC1-1 through PDSC1-8 by adding varying amounts of GRc and GRb3 to PDSC1
First, the mixture of GRc and GRb3 with high purity is prepared by separating the total saponins of the stem and leaf of pseudo-ginseng by a conventional method. Then, the mixture was added to PDSC1 to gradually increase the contents of GRc and GRb to obtain PDSC1 derivatives PDSC1-1 to PDSC1-8 compositions. The content of each of the ginsenoside and total saponin in these compositions was determined by HPLC analysis, and the ratio between them was calculated. The experimental results (tables 21 and 22) show that these compositions have a total saponin content above 92.8% and a ratio GRb/GRd that gradually increases from 1.03 to 1.75 for PDSC1, and gradually decreases from 2.17, 1.71 and 1.89 for PDSC1 to 0.91, 0.70 and 0.62 for GRb1/GRc, GRc/GRb3 and (GRb 1+ GRd)/(GRc + GRb 3), respectively, with a small variation in the ratio GRc/GRb3 (between 0.76 and 0.88).
Table 21 content of each of the ginsenoside and total saponins (%, n=3) in PDSC1 and its derivative PDSC compositions
Figure BDA0004007353830000312
Figure BDA0004007353830000321
TABLE 22PDSC1 and content configuration of the active ingredients of the respective panaxadiol saponins in the derivative PDSC compositions thereof
Figure BDA0004007353830000322
Example 7-2. Effect of higher ratio of GRc and GRb PDSC1 derivative compositions on the efficacy of treatment of PD.
The efficacy strength of the PDSC1 and the derivative thereof for relieving the PD motor symptoms is compared by using a HAL induced mouse stiffness model, and then the efficacy of the PDSC1 and the derivative thereof for resisting the ROT induced rat PD formation is compared and studied according to actual conditions.
As shown in Table 23, all six compositions PDSC1-1 to PDSC1-6 were very remarkable against the symptoms of stiff mice induced by HAL, and the potency was comparable to PDSC1. It can be seen that a substantial change in the ratio of GRb/GRd, GRb1/GRc, GRc/GRb3 and (GRb 1+ GRd)/(GRc + GRb 3) in PDSC1 does not affect the efficacy of PDSC1 against symptoms of PD exercise caused by insufficient dopamine signaling. At the same time, the results of the study also suggest the importance of the ratio GRc and GRb3 and GRc/GRb3 for the optimal composition against HAL-induced stiffness.
Table 23 increases the efficacy of PDSC1 derivative compositions at ratios GRc and GRb3 against HAL-induced stiffness in mice (mean±sem, n=10)
Figure BDA0004007353830000323
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p<0.001; compared with the HAL group after HAL administration, ### p<0.001。
however, as shown in Table 24, the four compositions PDSC1-1 through PDSC1-4 exhibited different strengths against ROT-induced formation of PD in rats and were not as potent as the parent PDSC1, with PDSC1-1 and PDSC1-3 being weaker and much less potent than PDSC1. The results of the study demonstrate that for PDSC1, while maintaining the GRc/GRb3 ratio substantially unchanged, increasing the GRb/GRd ratio and decreasing the GRb1/GRc, GRc/GRb3 and (GRb 1+ GRd)/(GRc + GRb 3) ratios simultaneously are detrimental to their efficacy against ROT-induced rat PD formation.
Table 24 effects of increasing ratio of GRc and GRb of PDSC1 derivative composition on Rotenone (ROT) induced rat PD formation (mean±sem, n=7)
Figure BDA0004007353830000331
Note that: experimental data were analyzed using a one-way analysis of variance method. In comparison with the normal group, *** p<0.001; in comparison with the set of ROT s, # p<0.05, ## p<0.01, ### p<0.001。
however, the comparative Table 20 shows that the 4 ratios of the best-efficacy compositions (very significant efficacy for both HAL and ROT models) are GRb1/GRd between 0.79 and 1.53, GRb1/GRc between 0.76 and 2.17, GRb1/GRb3 between 1.11 and 2.20, (GRb 1+ GRd)/(GRc + GRb 3) between 0.76 and 1.92, respectively, while the three ratios GRb1/GRc, GRb1/GRb and (GRb 1+ GRd)/(GRc + GRb 3) of the four PDSC1-1 to PDSC1-4 are all distributed within the best-efficacy ranges, and the ratio (1.52 to 1.66) of GRb1/GRd is also very close to the best-efficacy ranges (Table 22). This therefore illustrates that PDSC1 and other preferred PDSCs and HGCs have their specific ratios of GRb1/GRd, GRb1/GRc, GRb1/GRb3, and (GRb 1+ GRd)/(GRc + GRb 3).
Example 7-3. Significant increases in the GRc/GRb ratio affect the efficacy of a preferred composition in treating PD.
To further explore whether each of the compositions had its specific GRc/GRb3 ratio, based on the known compositions having a GRc/GRb3 ratio in the range of 0.79 to 2.11 (Table 15), the applicant used the composition PDSC9 as an example, changed the GRc/GRb3 ratio in the optimum range while maintaining the other 4 ratios substantially unchanged, and determined the effect of this change on the efficacy using the two animal models described above. To this end, the PDSC9-1 composition was constructed with a single ingredient to raise the GRc/GRb ratio from 0.92 to 1.68, keeping the other ratios substantially unchanged or less changed, with reference to the PDSC9, which is a preferred composition (Table 25).
TABLE 25 ginsenoside content configuration in PDSC9 and its derivative PDSC9-1
Figure BDA0004007353830000332
The efficacy of PDSC9 and PDSC9-1 was measured in the same manner as described above, and as shown in table 26, PDSC9-1 was found to be significantly resistant to HAL-induced stiffness symptoms in mice, but showed a decrease in efficacy intensity compared to PDSC 9.
Table 26 efficacy of PDSC9 derivative compositions with increased GRc/GRb3 ratio PDSC9-1 against HAL induced stiffness in mice (mean+ -SEM, n=10)
Figure BDA0004007353830000341
Note that: method for using single factor analysis of varianceExperimental data were analyzed. Compared to the HAL group prior to HAL administration, *** p<0.001; compared with the HAL group after HAL administration, ### p<0.001。
however, the potency of PDSC9-1 against ROT-induced rat PD formation was significantly weaker than that of PDSC9 (table 27). It can be seen that increasing the ratio GRc/GRb3 alone (from 0.92 to 1.68) in the 5 ratio in PDSC9 significantly reduces the potency. Looking back at Table 15, the ratio GRc/GRb3 of the most effective composition is distributed between 0.79 and 1.77. Therefore, the results of the study demonstrate that PDSC9 and other good PDSC and HGC also have their specific GRc/GRb3 ratios.
Summary and discussion of the results of the study of examples 3 to 7:
the results of the above studies in examples 3 to 7 show that both the preferred panaxadiol saponin compositions (APDSC) and the preferred holographic ginsenoside compositions (AHGC) are in a specific preferred compatible form (Table 15 and Table 17), or that each APDSC and AHGC is a separate organism. Further, 5 ratios of GRb1/GRd, GRb1/GRc, GRb1/GRb3, GRc/RRb3 and (GRb 1+ GRd)/(GRc + GRb 3) together form compatible parenchyma of AHGC and APDSC. Specifically, the ratio of HGC1 to PDSC1 is 1.03, 2.17, 1.71, 0.79 and 1.89, the ratio of HGC2 to PDSC2 is 0.79, 1.38, 2.20, 1.59 and 1.92, the ratio of HGC3 to PDSC3 is 1.56, 1.00, 2.11 and 1.11, the ratio of HGC4 to PDSC4 is 1.45, 0.76, 1.11, 1.46 and 0.76, the ratio of HGC5 to PDSC5 is 1.13, 0.75, 1.32, 1.77 and 0.90, the ratio of HGC9 to PDSC9 is 1.53, 1.39, 1.28, 0.92 and 1.10, the ratio of HGC10 to PDSC10 is 1.39, 1.01, 0.93, 0.92 and 0.83, and these 5 ratios have respective upper and lower limits of 1.56 and 0.79 (GRb 1/GRd), 2.17 and 0.75 (GRb 1/GRc), 2.20 and 0.93 (GRb 1/GRb 3), 2.11 and 0.79 (GRc/GRb 3), 1.92 and 0.76 (GRb 1+ GRd)/(GRc + GRb), respectively. However, the ratios of TPDS/TPTS, re/Rg1 and Rb1/Re in these AHGC may vary within a range of from 1.88 to 4.29, from 2.33 to 4.03 and from 0.64 to 1.53, respectively, and it is not precluded that these ratios extend in greater or lesser directions. The ratio parameters provide the basis for preparing the active or effective holographic ginsenoside composition (AHGC) and the active and effective panaxadiol saponin composition (APDSC) by comprehensively utilizing the ginseng medicinal materials, and also lay a solid foundation for the subsequent research of the internal connection between the effective component compatibility and the medicinal effect.
Table 27 Effect of PDSC9 derivative composition of increasing the ratio GRc/GRb3 PDSC9-1 on Rotenone (ROT) induced rat PD formation (mean+ -SEM)
Figure BDA0004007353830000342
Note that: experimental data were analyzed using a one-way analysis of variance method. In comparison with the normal group, *** p<0.001; in comparison with the set of ROT s, # p<0.05, ## p<0.01, ### p<0.001; in comparison with the PDSC9 group, & p<0.05。
clearly, the discovery of these independent organisms (APDSC and AHGC) is not a clear reasoning result, but rather a scientific discovery of anti-routine knowledge, a significant application and theoretical value. The method provides scientific basis for the establishment of product quality standards, can effectively relieve the shortage of ginseng and American ginseng resources, and breaks the knowledge of traditional Chinese medicine on the medicinal value, economic value and use mode of ginseng, american ginseng and pseudo-ginseng and stems and leaves thereof. The root and stem leaves are used together, so that the consumption of precious root medicinal materials is saved, the medicinal value of the low-cost stem leaves is exerted, the preparation cost of the optimal composition is greatly reduced, and the raw medicinal material resources for popularization and application of subsequent products are greatly widened. Particularly, the excellent holographic ginsenoside composition (AHGC) and the excellent panaxadiol saponin composition (APDSC) prepared by the compatibility of the root and the stem and the leaf generate the drug effect which can not be generated by the ginsenoside composition or the mixture prepared by the root or the stem and the leaf alone, or the effect of the AHGC and the APDSC on the treatment of both the symptoms and root causes of PD breaks through the traditional medicinal value of ginseng, american ginseng and pseudo-ginseng. More importantly, AHGC and APDSC can effectively break the dead bureau of medicaments for treating both principal and secondary aspect of PD at home and abroad for a long time.
It is also particularly pointed out that the research results also show that each optimal composition contains complex and mutually synergistic effects or just partial work cooperation among the ginsenoside GRb, GRd, GRc and GRb3, so that a synergistic effect and even new pharmacological effects and medicinal effects are generated to play a role in treating both symptoms and root causes of PD. Obviously, the complex and mutually synergistic or separately cooperative relationship is further revealed, so that the pharmacological connotation of the compatibility between the active ingredients in the AHGC and the APDSC can be further deeply understood, and the method has important significance in further excavating the medicinal value of the ginsenoside. Accordingly, the applicant conducted a study of example 8.
Example 8. The pharmacodynamic contributions of different functional units in a super-effective composition to treat both principal and secondary aspect of the super-effective composition to PD are studied to reveal the potential complex interactions or just the job-division cooperation between the different active ingredients.
According to the two functional units in the optimal composition, the drug effect of treating both principal and secondary aspect of PD of the optimal composition is studied by taking PDSC1 as an example, so as to verify that the two functional units have a division and cooperation relationship.
Example 8-1. Effect of functional units (GRb 1+ GRd) and (Rc+Rb3) on PDSC1 to improve HAL-induced stiffness symptoms.
The research method comprises the following steps: the test groups and dose designs are shown in Table 28. 2 dose groups according to the optimum dose of PDSC1 of 60mg/kg and designed functional units (GRb 1+ GRd) and (GRc + GRb 3): (1) 60mg/kg (hereafter referred to as full dose), which would be an indication of whether the two functional units have full composition-like effects; (2) the absolute doses (hereafter constituted doses) in the case of 60mg/kg PDSC1, respectively, namely 36mg/kg (GRb 1+ GRd) and 24mg/kg (GRc + GRb 3), are set forth to clarify the relative contributions of the two functional units in the palliative treatment.
Table 28 effect of split compositions of PDSC1 on HAL-induced mouse stiffness (mean±sem, n=10
Figure BDA0004007353830000351
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p<0.001; compared with the HAL group after HAL administration, # p<0.05, ## p<0.01, ### p<0.001; in comparison with the PDSC1 group, + p<0.05, ++ p<0.01,+++p<0.001。
study results: as shown in Table 28, at 60mg/kg, the split composition (GRb 1+ GRd) significantly shortened the 2 indicators reflecting stiffness, but the strength of action was less than that of the full composition PDSC1, while the functional unit (GRc + GRb 3) did not improve the symptoms of stiffness. Interestingly, at the actual constituted dose of PDSC1, (1) (GRb 1+ GRd) (36 mg/kg) aggravates instead both HAL-induced stiffness symptoms and (2) (GRc + GRb 3) (24 mg/kg) fail to ameliorate HAL-induced stiffness symptoms, but the protective efficacy of PDSC1 can be fully reproduced when the two are combined ((1) + (2)) to reconstruct the compositional configuration of PDSC 1. It can be seen that neither the split fractions (GRb 1+ GRd) nor (GRc + GRb 3) can replace the full composition PDSC1, and that the effect of (GRb 1+ GRd) on improving the symptoms of stiffness is superior to that of (GRc + GRb 3), but the effective dose range is narrow and the dose-effect relationship shows a jumping bidirectional change. Although (GRc + GRb 3) is insufficient for improving stiff symptoms, the drug can exert positive efficacy in cooperation with (GRb 1+ GRd), and simultaneously the side effects of (GRb 1+ GRd) can be restrained, so that the overall efficacy is remarkable, stable and reliable.
The results of the study verify the correctness of the division of the two functional units by the applicant in terms of efficacy and reveal the proper division of work between the two functional units in the total alleviation of stiff symptoms. In particular, the results of the study demonstrate that the efficacy of the whole recipe in alleviating the symptoms of stiffness is a consequence of a qualitative change in the amount of change produced by the appropriate division of work between the two functional units.
Example 8-2. The quantitative relationship of functional units (GRb 1+ GRd) was explored.
The unusual dose and pharmacodynamic relationship observed in example 8-1 for the functional unit (GRb 1+ GRd) fraction, namely significant efficacy at 60mg/kg, but not only at 36mg/kg, was not effective but rather significantly aggravated HAL-induced rigidity in mice. The dose-effect relationship does not accord with the conventional knowledge of the expert on the dose-effect relationship, namely the dose-effect relationship of traditional Chinese medicines is often expressed as an inverted U shape: the efficacy increases with increasing doses over a range, followed by a plateau and then a decrease in efficacy. Therefore, it is intended to further confirm this seemingly contradictory result and further observe whether there is a low dose (GRb 1+ GRd) that aggravates stiffness and further reduce the dose to 24mg/kg, which is the absolute amount of (GRb 1+ GRd) contained in 40mg/kg PDSC 1.
Table 29 dose influence functional unit (rb1+rd) effect on HAL-induced mouse stiffness (mean±sem, n=10)
Figure BDA0004007353830000361
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p<0.001; compared with the HAL group after HAL administration, ## p<0.01, ### p<0.001。
as shown in Table 29, (GRb 1+ GRd) the test results of example 8-1 were repeated at doses of 60mg/kg and 36 mg/kg. Unexpectedly, however, the 24mg/kg dose (GRb 1+ GRd) significantly shortens the turn-around time (Tturn) and the total pole-climbing time (Ttotal), and the mathematical averages thereof show that the potency of this dose tends to exceed that of the high dose group. It can be seen that the dose-response relationship of (GRb 1+ GRd) presents a high dose to relieve stiffness, a medium dose to exacerbate stiffness, and a low dose to be effective as a non-inertial jump. The findings of example 8-1, not only confirm that the dose range for alleviating the symptoms of stiffness is narrow (GRb 1+ GRd), the dose-response relationship exhibits a two-way jump change, and further that such two-way jump change can occur at both ends of the dose that produces the side effects.
Examples 8-3. Individual GRb1 and GRd contributed to the study of the HAL-induced stiffness alleviation by PDSC 1.
On the basis of defining the contribution of the functional unit (GRb 1+ GRd) to the relief of stiff symptoms of the full composition PDSC1, (GRb 1+ GRd) was split into individual components to elucidate the pharmacodynamic contribution of GRb1 and GRd to the functional unit (GRb 1+ GRd). The results of the experiment are shown in Table 30, and at a dose of 60mg/kg, each of the individual GRb and GRd showed partial efficacy, but the efficacy was not as strong as the efficacy of the functional unit (GRb 1+ GRd) and was mainly expressed as: the efficacy was close to the turn-around time (Tturn) at the tip of the rod, GRb1 and functional unit (GRb 1+ GRd), whereas GRd did not show significant efficacy; in contrast, for the total pole-climbing time (Ttotal), the potency of GRd was close to that of the functional unit (GRb 1+ GRd), whereas GRb1 did not show significant potency. The results of the study are fully indicative that GRb and GRd are active ingredients for alleviating the symptoms of stiffness by functional units (GRb 1+ GRd), and cannot be replaced by each other, and must be used together.
Contributions of Table 30, individual GRb and GRd to functional units (GRb 1+ GRb) to alleviating the effects of HAL on mouse stiffness induction (mean+ -SEM, n=10)
Figure BDA0004007353830000362
/>
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p<0.001; compared with the HAL group after HAL administration, # p<0.05, ### p<0.001。
examples 8-4 illustrate that GRc and GRb are both active ingredients of a functional unit (GRc + GRb 3).
Based on the experimental results described above, experiments were designed here to further understand the relative contribution of individual GRc and GRb3 to the functional unit (GRc + GRb 3). For this reason, the comparative observation of whether (GRc + GRb 3) and (GRb 2+ GRb 3) can reverse the effect of 36mg/kg dose of (Rb 1 +Rd) to exacerbate stiffness was a remission effect and an intensity of effect. According to the content ratio of individual ginsenosides in PDSC1, when the dose of PDSC1 is 60mg/kg, the dose of the (GRb 1+ GRd) fraction is 36mg/kg, the dose of (GRc + GRb 3) fraction is 16.3mg/kg, and the dose of (GRb 2+ GRb 3) fraction is 12.4mg/kg. Accordingly, the experimental group and the dose design are shown in table 31, and in order to exclude the influence of non-investigation factors on the experimental results, the PDSC1 control group of the corresponding dose was also set in the experiment.
The experimental results are shown in Table 31, in which the dose of 60mg/kg of PDSC1 significantly shortens the two parameters reflecting stiffness compared to the HAL model, while the dose of 36mg/kg (GRb 1+ GRd) significantly increases the turn-around time and pole-climbing time, again indicating that the effective dose of PDSC1 (GRb 1+ GRd) significantly exacerbates the symptoms of stiffness induced by HAL when used independently from (GRc + GRb 3). However, the potency of the individual ginsenosides in the total composition was similar to that of PDSC1 ((2)) having only a deficiency of GRb2, which was constituted by (GRc + GRb 3), was added to the composition in a dose of 36mg/kg (GRb 1+ GRd), and it was explained again that GRb did not greatly contribute to the potency of the composition (example 5-1); while the addition of (GRb 2+ GRb 3) of the resulting PDSC1 ((3)) lacking GRc significantly reduced pole climbing time, the mathematical average was 1.43 times that of the GRc-containing composition, and failed to significantly reduce turn around time. These findings again demonstrate that functional unit (GRc + GRb 3) can convert the efficacy of functional unit (GRb 1+ GRd) from exacerbating stiffness to a protective efficacy that relieves stiffness, and further indicate that both GRc and GRb3 are active ingredients of functional units.
From the above results, it can be seen that the two functional units (GRb 1+ GRd) and (GRc + GRb 3) are slightly effective and ineffective when used independently, even aggravate symptoms, and the drug effect is remarkable, stable and reliable when used simultaneously, which means that the drug effect when the two functional units are combined is not only variable but also is not necessary for the PDSC1 to be effective against PD exercise symptoms. Moreover, the qualitative change of the drug effect further demonstrates the action characteristics of the proper division of work of the two functional units in the optimal effect composition.
Contribution of table 31GRc and GRb3 to alleviating the efficacy of HAL-induced stiffness in functional units (GRc + GRb 3) alleviating the contribution of HAL-induced stiffness in mice (mean±sem, n=10)
Figure BDA0004007353830000371
Note that: experimental data were analyzed using a one-way analysis of variance method. Compared to the HAL group prior to HAL administration, *** p<0.001; compared with the HAL group after HAL administration, # p<0.05, ## p<0.01; in comparison with the PDSC1 of the present invention, +++ p<0.001。
results of the study of examples 8-1 to 8-4 are summarized and discussed:
the ginsenoside GRb, GRc, GRb3 and GRd are all effective components for alleviating the symptoms of PD stiffness in the ginsenoside composition PDSC1, but any one of two functional units (GRb 1+ GRd) and (GRc + GRb 3) consisting of the effective components cannot represent the whole composition for alleviating the symptoms of PD stiffness, and subtle division cooperation (also called just division cooperation) of the two components brings the special advantages of strong efficacy, stability, reliability, high safety and wide effective dosage range of the whole PDSC 1. This subtle division of work is achieved as if, (GRb 1+ GRd) is a direct practitioner for alleviating stiffness, but its effective dose range is narrow, and the dose-response relationship (high dose effective, medium dose exacerbating stiffness, low dose effective) exhibits reverse conventional skip bidirectional changes, and therefore cannot be dosed alone; while (GRc + GRb 3) appears to be a coordinator, not only can the synergistic effect (GRb 1+ GRd) of the drug effect of relieving stiffness be generated, but also the drug effect of exacerbating stiffness can be restrained, and the jumping type bidirectional dose-effect relationship is converted into a stable forward dose-effect relationship, so that the effective dose range (60 mg/kg to 80 mg/kg) of the PDSC1 is greatly expanded.
Furthermore, the subtle partnership of these two functional units also fully explained that, although there was a large difference in the content of GRb1, GRc, GRb3 and GRd in the holographic ginsenoside compositions (HGC 1 to HGC 10) and the corresponding panaxadiol saponin compositions (PDSC 1 to PDSC 10), they were both significantly resistant to HAL-induced stiffness symptoms in mice (tables 15 and 18).
Examples 8-5. The contributions of functional units (GRb 1+ GRd) and (GRc + GRb 3) to PDSC1 in controlling PD formation were investigated.
On the basis of defining the significance of the compatibility of the two functional units (GRb & lt1+ & gt GRd) and (GRc & lt GRb3 & gt) for the optimal composition to relieve the PD exercise symptoms, the compatibility significance of the two functional units for preventing and treating PD formation (root cause) of the PDSC1 is studied by using a model of the ROT-induced rat PD capable of reflecting the root cause effect.
The test groups and dose designs are shown in Table 32. According to the best dose of PDSC1 to the present model of 40mg/kg, 2 dose groups of functional units (GRb 1+ GRd) and (GRc + GRb 3) were designed: (1) 40mg/kg, which is an amount that demonstrates whether or not any of the functional units has a full composition-like effect; (2) absolute doses of each of 40mg/kg PDSC1, namely a 24mg/kg dose (GRb 1+ GRd) and a 16mg/kg dose (GRc +rb3), respectively, which illustrate the relative contribution of either functional unit in the efficacy of preventing PD formation (root cause control); the PDSC1 control group was also set. The methods for the ROT-induced rat PD model, dosing and PD clinical behavior scoring, and quantitative determination of limb movements (forelimb stride test and forelimb lift test) and coordination balance ability (rotarod test) were the same as above.
Experimental results (table 32) indicate that PDSC1 can significantly combat the disease changes of each observed index induced by ROT at a dose of 40mg/kg, with some parameters approaching normal levels; none of the functional units (GRb 1+ GRd) showed any protective effect at a dose of 40mg/kg, some of which were numerically even inferior to the model group, but some of which were mathematically superior to the model at a low dose of 24mg/kg, especially the residence time on the rotor bar was significantly longer than in the high dose and model groups, so it could be considered that the actual dose in the full composition (GRb 1+ GRd) was partially resistant to ROT-induced PD formation. Remarkably, the pharmacodynamic behavior of the functional unit (GRc +rb2+ GRb 3) is significantly different from that of the functional unit (GRb 1+ GRd). (GRc + GRb + GRb 3) at 40mg/kg, all indices are only mathematically superior to the model group; while 16mg/kg could significantly combat ROT-induced rat PD formation, none of the four study indicators were as mathematical as the PDSC1 group, with a significantly shorter residence time on the rotor bar than the PDSC1 group. The above study results demonstrate that: (1) Any functional unit is far from representing a full composition to combat ROT-induced PD formation and development; (2) (GRc +Rb3) has a stronger activity against ROT-induced PD formation and development than (GRb 1+ GRd), but the effective dose range is narrow, even in a punctiform distribution, which is essentially different from the full composition effective dose range of 30-60 mg/kg (example 3); (3) (GRb 1+ GRd) alone, depending on the dosage or exacerbation of PD development or exhibiting weak protective efficacy, but the formation of PDSC1 together with (GRc + GRb 3) not only synergistically results in a stronger efficacy, but also greatly expands the effective dosage range.
TABLE 32 effects of functional units (GRb 1+ GRd) and (GRc + GRb 3) on PDSC1 control of PD formation (mean+ -SEM)
Figure BDA0004007353830000381
Note that: experimental data were analyzed using a one-way analysis of variance method. In comparison with the normal group, *** p<0.001; in comparison with the set of ROT s, # p<0.05, ## p<0.01, ### p<0.001; in comparison with the PDSC1 group, & p<0.05。
it can be seen that, for PDSC1, there is a subtle or just-right partnership between (GRb 1+ GRd) and (GRc + GRb 3) for preventing and treating PD disease, but contrary to alleviating stiff symptoms, (GRc + GRb 3) appears to be a direct implementation of drug efficacy, while (GRb 1+ GRd) is a coordinator, but it is needless to say that the mutual cooperation of the two components not only eliminates the respective drug efficacy defect but also produces a brand-new drug efficacy behavior, so that the drug efficacy of the whole prescription (PDSC 1) is strong, stable and reliable, highly safe, and has a wide effective dosage range.
Summary and discussion of the results of the study of examples 8-1 to 8-5:
from the above results of the study in example 8, it was further verified that GRb, GRc, GRb3 and GRd, which were found by the applicant before, are the main active ingredients of Active Holographic Ginsenoside (AHGC) and active panaxadiol saponin composition (APDSC), respectively, and that the scientificity of dividing these 4 individual panaxadiol saponin ingredients into two functional units was also verified from the efficacy. In particular, a subtle division cooperation relationship of the two functional units in the process of treating both symptoms and root causes of PD in AHGC and APDSC is disclosed. For the treatment of the symptoms, (GRb 1+ GRd) seems to be a direct implementation of the drug effect, while (GRc + GRb 3) is a coordinator; for root cause, their roles are reversed. The two components have a separate work and cooperation relationship, and the two components are mutually matched to eliminate the respective drug effect defects and generate brand new drug effect behaviors no matter how the two components are indispensable for treating both symptoms and root causes, so that the full composition of the AHGC or the APDSC has strong drug effect, stability, reliability, high safety and wide effective dosage range. This in turn reasonably explains that the 5 ratios (GRb/GRd, GRb1/GRc, GRb1/GRb, GRc/GRb3 and (GRb 1+ GRd)/(GRc + GRb 3) found by the applicant in the foregoing collectively determine the potency, efficacy or inefficiency of the Holographic Ginsenoside Composition (HGC) and the panaxadiol composition (PDSC), and that each AHGC and the corresponding APDSC have their own independent 5 ratios, or that they are independent organisms of specific content configurations (GRb 1+ GRd) and (GRc + GRb 3) that treat both principal and secondary aspects of PD.
It is also pointed out here in particular that, at the point of view of the pathophysiology of the condition, the subtle partnership of functional units (GRb 1+ GRd) and (GRc + GRb 3) reflects their systemic treatment against the disease network, thus creating a rapid jump in the potency from quantitative to qualitative as compared to the single component and functional unit. Haloperidol (HAL) -induced stiffness is closely related to the inhibition of dopamine 2-type receptor-induced excessive release of striatum, especially acetylcholine. Mitochondrial complex enzyme I inhibitor ROT mimics almost all of the pathological mechanisms of human PD occurrence and development when induced in rodent PD models, including: mitochondrial dysfunction and its malignant biochemistry and cellular events such as ATP deficiency, oxidative stress injury, neuroinflammation, glial dysfunction, abnormal deposition of gamma-synuclein, and neurodegeneration. Accordingly, applicants will continue to take PDSC1 as an example in the following examples to verify the systemic therapeutic effect of a beneficial composition on a network of conditions.
Example 9. Study of the effective composition against PD and its cell parenchyma in neurodegenerative diseases.
Brain function and brain health depend on the structure and functional integrity of neurovascular units (NVU), and there is a direct or indirect link and interaction between the cell types of members in the unit, including vascular endothelial cells, astrocytes and neurons, on function and cell bodies, so that impaired function or structure of any one member may lead to lesions and related diseases of the unit. Indeed, dysfunction and structural destruction of NVU, including brain microvascular damage, blood brain barrier destruction, astrocyte dysfunction and neurodegeneration, are common pathological features of neurodegenerative diseases including Parkinson's Disease (PD), alzheimer's Disease (AD), multiple Sclerosis (MS) and amyotrophic lateral sclerosis (ALS, progressive freezing), and are intimately associated with the occurrence and progression of the disease. NVU dysfunction is also closely related to the occurrence of ischemic stroke and prognosis of malignancy. Mitochondrial dysfunction is thought to be a common pathological event in neurodegenerative diseases that can induce and exacerbate disease occurrence and progression. Therefore, the drug effect of the tested drug for protecting the neurovascular unit member cells against mitochondrial dysfunction can reflect the basic drug effect of the drug on PD and other neurodegenerative diseases from the aspects of the common mechanism and the neurovascular unit.
Therefore, the research on the effect of the ginsenoside optimal composition and the functional units thereof on protecting brain cells of each member of the neurovascular unit against ROT-induced mitochondrial dysfunction can reveal the potential of the optimal composition for preventing and treating the cell parenchyma of PD (treating the root cause of PD) and other neurodegenerative diseases and the proper division cooperation of two main functional units in the ginsenoside composition at the cell level, so that the scientific connotation of preventing and treating the PD and the development of the optimal composition is enriched and the disease range of preventing and treating the neurodegenerative diseases is enlarged.
According to the pharmacodynamic characteristics of the panaxadiol saponin composition PDSC1 for preventing and treating the formation of the ROT-induced PD rat model and the subtle/proper division of work of two functional units, the applicant believes that the PDSC1 can protect each member cell of NVU, and the cell types of which the two functional units are preferentially protected can be different. However, it is not known how different member cells interact in NVU during the occurrence and development of PD and other neurodegenerative diseases, such as who affects other member cells after disease, etc., so that it is impossible to suppress diseases in early stages or sprouting states by precisely targeting upstream pathological events or the most fragile member cells, and it is also impossible to accurately understand the mechanism of action of the optimal composition, particularly the division of work where the functional unit is right, in the division of work at the neurovascular unit level.
Therefore, the applicant will first compare the vulnerability of the main three member cell types of NVU to ROT inhibiting mitochondrial complex I to reveal the most fragile member cells that may be present in the unit; then, exploring whether the most fragile cells release inflammatory factors or toxic substances after being damaged or diseased, thereby reducing the toxicity of peripheral cells against the loss of complex enzyme I functions; finally, the protective effect of PDSC1 and its functional unit on neurovascular unit and its action mechanism are studied.
Example 9-1. Comparative study of vulnerability of three member cells of NVU to ROT inhibiting mitochondrial Complex enzyme I and protection of PDSC 1.
The vulnerability of three member cells of NVU to ROT inhibiting mitochondrial complex I was studied in comparison to reveal the most vulnerable member cells that may be present in the unit. Human Brain Microvascular Endothelial Cells (HBMEC), rat primary Astrocytes (ASC) and dopamine neuron model hyperdifferentiated PC12 cells were cultured by conventional methods. After 24h plating, mitochondrial complex enzyme I inhibitor ROT solutions of different concentrations, each 5-well (n=5), were added to the culture medium, and then the culture was continued for 72h, and the cell viability was measured by the sulfonylrhodamine B (SRB) method and the inhibition (%) of ROT on cell viability was calculated. As shown in table 33, ROT reduces the cell viability of endothelial cells, astrocytes and dopamine neurons in a concentration-dependent manner.
TABLE 33 Activity of Rotenone (ROT) against 3 member cells of neurovascular unit (NVU) (%)
Figure BDA0004007353830000391
Figure BDA0004007353830000401
Comparison with normal control: * p <0.05, < p <0.01, < p <0.001; n=5.
Calculating IC of ROT to different cells by SPSS.20 software according to the quantitative effect relationship data of each cell 50 The (half inhibition concentration) values are respectively: human Brain Microvascular Endothelial Cells (HBMEC) were 230.56nM; astrocytes (ASC) were 1980nM; dopamine neuron (PC 12) is549.33 (Table 34).
The research results show that, in the neurovascular unit, the brain microvascular endothelial cells are most sensitive/fragile to the loss of mitochondrial complex enzyme I, and next to neurons, astrocytes have strong capability of coping with the loss of mitochondrial complex enzyme I. The research result is consistent with the common cerebral microvascular injury of PD patients, and shows that protecting cerebral vascular endothelial cells has important therapeutic value for preventing and treating PD. Particularly, the research results provide a brand-new clue for the subsequent disclosure of the interaction of each member cell in the neurovascular unit on the development of PD, and provide clues for the subsequent further deep disclosure of the principle of the ginsenoside optimal composition for treating both the symptoms and root causes of PD and the scientificity of the interaction of two functional units and the content configuration of the effective components.
TABLE 34 half Inhibition Concentration (IC) of rotenone on 3 member cells of neurovascular unit (NVU) 50 )(n=5)
Figure BDA0004007353830000402
Next, the effect of the panaxadiol saponin optimal composition PDSC1 on protecting each member cell in the neurovascular unit against the decrease in cell viability by the mitochondrial complex enzyme I inhibitor ROT was observed to confirm the consistency or difference in the protective effect of the optimal composition on each member cell. Culturing the test cells by conventional methods, and culturing the test cells by using the IC of the ROT of each member cell 50 The concentration of the values was treated for 48 hours to induce cell damage, PDSC1 was applied at different concentrations, respectively, before the ROT was applied, while normal and ROT controls were set, and cell viability was measured using SRB.
Table 35 optimal compositions PDSC1 protects neurovascular unit member cells against rotenone toxicity
Figure BDA0004007353830000403
Comparison with ROT control: * p <0.05, < p <0.01, < p <0.001, n=5.
As shown in Table 35, PDSC1 significantly protects Human Brain Microvascular Endothelial Cells (HBMEC), astrocytes (ASC) and dopamine neuron PC12 cells against ROT-induced cell damage at concentrations ranging from 1.25 to 55.0. Mu.M, with no significant difference between the individual member cells at the optimal concentrations ranging from 1.25 to 25. Mu.M. It can be seen that the optimal composition can protect each member cell in the neurovascular unit including the most fragile brain microvascular endothelial cells against cytotoxicity induced by low or absent mitochondrial complex enzyme I function, and has a wide range of effective concentrations. Mitochondrial complex enzyme I dysfunction or loss is an important risk factor for PD and mitochondrial dysfunction is a significant cause of neurodegeneration. The results of the study strongly support the palliative effect of the optimal composition against the development and progression of diseases induced by risk factors.
Next, the effect of PDSC1 on anti-ROT-induced HBMEC apoptosis was further observed at a concentration of 25 μm. Apoptosis rates were determined using conventional commercial kits. As shown in table 36, ROT (100 nM) treatment for 48 hours induced apoptosis in nearly 30%; PDSC1 not only can reduce the apoptosis rate of cells in basal state (compared with normal control group), but also can significantly combat ROT-induced endothelial apoptosis. The results of the study demonstrate the safety and efficacy of PDSC1 to protect vascular endothelial cells against PD risk factors (mitochondrial complex I insufficiency).
Table 36 Effect of panaxadiol saponins with respect to PDSC1 in resisting ROT-induced apoptosis of HBMEC
Figure BDA0004007353830000411
HBMEC, human brain microvascular endothelial cells; PDSC1 panaxadiol saponin composition 1; ROT: rotenone; *** p<0.001 (vs. normal control group); ### p<0.001 (vs. ROT group); n=5.
Example 9-2. Ginsenoside-optimized composition protects the relationship between the pharmacological actions of neurovascular units and the effects of preventing and treating the occurrence and development of PD.
In order to further elucidate the relationship between the pharmacological effect of the ginsenoside-based potent and potent composition for protecting neurovascular units and the effect thereof for controlling the occurrence and development of PD, the potent and potent effects of the potent and potent composition on ROT-induced rat PD model were particularly compared and observed herein to protect neurovascular units against ROT cytotoxicity. The experimental data (table 37) show that the effect of the optimal, effective and ineffective compositions on protecting Human Brain Microvascular Endothelial Cells (HBMEC), astrocytes (ASC) and dopaminergic neuropc 12 cells were not significantly different nor were the total protection rates for the various cell types significantly different.
Table 37 holographic panaxadiol saponins protecting neurovascular unit member cells against ROT toxicity
Figure BDA0004007353830000412
Comparison to the normal control group: * P < 0.001; comparison to the ROT group: ### p<0.001;n=5。
since the malignant cycle formed by the interactions between the members of the neurovascular unit leads to the development of PD induced by the insufficient function of mitochondrial complex enzyme I, uniform protection of each member cell may be a key to determine whether Holographic Ginsenoside Composition (HGC) and corresponding panaxadiol saponin composition (PDSC) are optimally effective. Accordingly, the total protection rate (%) of each ginsenoside composition to the three member cells, the difference between the three, and the relationship between the sum of the differences (abbreviated as protection discreteness) and the overall efficacy are further analyzed. As shown in table 38, the optimal composition has the most uniform protective effect on the three member cells, and is characterized by the minimum difference between the maximum protective rate and the minimum protective rate, the smaller difference between the middle value and the two end values, and the minimum protective discreteness; the variability of the protective rate of the ineffective composition for the three member cells is greatest, while the variability of the protective rate of the effective composition for the three member cells is in between. In particular, the ineffective composition PDSC1-1 has the strongest protective effect on vascular endothelial cells (HBMEC), while the effective composition PDSC1-2 has the stronger effect on protecting vascular endothelial cells (HBMEC) and Astrocytes (ASC) than the effective composition PDSC1, but has weaker effect on nerve PC12 cells than the effective composition, but both effective compositions PDSC1-2 and PDSC1-4 can uniformly protect vascular endothelial cells (HBMEC) and Astrocytes (ASC), and since astrocytes have the function of protecting nerves, both cell types can be simultaneously protected to be neuroprotective under the whole condition.
Table 38 relationship between overall potency and cytoprotective effects on members of the neurovascular unit
Figure BDA0004007353830000413
/>
Figure BDA0004007353830000421
In conclusion, the research results show that the uniform protection of each member cell type in the neurovascular unit is important for preventing and controlling the generation and development of PD induced by mitochondrial complex enzyme I function deficiency, and the optimal composition can uniformly protect each member cell. In contrast to the five ratios of these optimal compositions (PDSC 1) and their PDSC 1-derived compositions (PDSC 1-1 to PDSC 1-6) and the efficacy against ROT-induced formation of PD in rats, it can be seen that subtle changes in the five ratios can bring about shifts in the protection of the neurovascular unit member cell types, thereby affecting the intensity or presence of the overall efficacy. This further reveals that the five ratio of the optimal composition converts to mysterious which prevents the onset and progression of the ROT-induced rat PD model.
Example 9-3 protection of neurovascular unit member cells by functional units (GRb 1+ GRd) and (GRc + GRb 3).
For PDSC1, the single component was used to combine the recombinant PDSC1 with its two functional units (GRb 1+ GRd) and (GRc + GRb 3). The PD cell model of Human Brain Microvascular Endothelial Cells (HBMEC), rat primary Astrocytes (ASC) and dopamine nerve P12 cells induced by Rotenone (ROT) was compared to observe the effects of PDSC1, reconstituted PDSC1, two functional units (at the same dose as the whole composition and the actual dose in the composition, respectively) and the combination of the two functional units at the actual dose, and the study results revealed the difference between the whole composition and the functional units and between the functional units. The test dose of PDSC1 was set to 25 μm; PDSC1, the functional units, were constructed with a single component based on the 5 components of PDSC1 at 28.14% grb1, 12.95% grc, 8.02% grb2, 16.48% grb3, and 27.44% rd, respectively, in total panaxadiol saponins (table 39). Cell culture, drug treatment and cell viability assays were as before.
As shown in table 39, the recombinant PDSC1 and the original PDSC1 combined with four individual ginsenosides GRb, GRc, GRb3 and GRd were able to significantly combat Rotenone (ROT) -induced reduced viability of Human Brain Microvascular Endothelial Cells (HBMEC), rat primary Astrocytes (ASC) and dopamine nerve P12 cells, and the potency was the same, indicating that these four ginsenosides could reconstruct the entire activity of PDSC 1.
TABLE 39 action of PDSC1 and its functional units against rotenone-induced decrease in cell viability
Figure BDA0004007353830000422
Figure BDA0004007353830000431
Comparison to the normal control group: * P < 0.001; comparison to the ROT model group: # p<0.05, ## p<0.01, ### p<0.001; comparison to PDSC1 group: & p<0.05;n=5。
in particular, studies have found that: at the same dose (25. Mu.M) as the whole composition and at the actual dose (13.9. Mu.M) in the composition, the functional units (GRb 1+Rd) can obviously protect vascular endothelial cells (HBMEC), astrocytes (ASC) and dopamine nerve P12 cells, but the action intensity is obviously lower than that of the whole composition PDSC1 and recombinant PDSC1; under the corresponding conditions, (GRc + GRb 3) can also protect the three cells obviously, but the efficacy strength is obviously lower than that of the full-composition PDSC1 and the recombinant PDSC1, however, the protection effect strength of the two functional units on the three member cells is the same as that of the PDSC 1. These study data demonstrate that (GRb 1+ GRd) and (GRc + GRb 3) are functional units of PDSC1 and none replace the full composition.
Of particular note, the study data further indicated: the protection intensity of (GRb 1+ GRd) on Astrocytes (ASC) is better than (GRc + GRb 3) under both dosage conditions, and the drug effect is close to that of the whole composition at the actual dosage (13.9 mu M) in the whole composition; the effect on protecting vascular endothelial cells (HBMEC) is the opposite, namely (GRc + GRb 3) has better efficacy than (GRb 1+ GRd) and the efficacy is close to that of the whole composition at the actual dose (7.36. Mu.M) in the whole composition; however, the protective effect on dopaminergic neuropcs 12 cells at both concentrations (GRc + GRb 3) has an advantage over (GRb 1+ GRd), whereas the combination of these two functional units fully reproduces the potency of the full composition at the actual concentrations in the full composition. These results demonstrate that (GRb 1+ GRd) and (GRc + GRb 3) are the main active ingredients of PDSC1 for protecting PD astrocytes and vascular endothelial cells, respectively, and that the latter protect dopamine nerve cells more strongly, especially both can synergistically protect individual member cells in the neurovascular unit. Thus, when combined in the appropriate ratio, the individual member cells of the neurovascular unit are maximally protected.
In summary, the above results show that the protecting effect on neurovascular units, two different functional units exist in the panaxadiol saponin composition PDSC1, and the common effect is more weight of each division; the common feature is that it has protective effect on three member cells of neurovascular unit, but the function unit (GR1+ GRd) has prominent effect of protecting Astrocyte (ASC), while the function unit (GRc + GRb 3) has stronger protective effect on cerebrovascular endothelial cells (HBMEC) and dopamine neurons. Therefore, the optimal composition ensures that the effective components and the content are configured to ensure that the effective components and the content can protect each member cell in the neurovascular unit to the greatest extent.
Results of associative PD animal models: (GRb 1+ GRd) is the main active ingredient against the motor symptoms caused by dopamine signal deficiency, and (GRc + GRb 3) is the main active ingredient against Rotenone (ROT) induced PD formation and development, suggesting that Astrocyte (ASC) dysfunction is associated with PD motor symptoms, whereas brain microvascular endothelial cell (HBMED) dysfunction or degeneration is closely associated with PD formation and development. Indeed, astrocytes are critical in maintaining the striatal extracellular neurotransmitter, particularly the excitatory neurotransmitter glutamate, and striatal neurotransmitter disorders are closely related to PD motor and non-motor symptomatic expression; neural function and structural degeneration, astrocyte dysfunction and brain microvascular destruction are common pathological mechanisms for the occurrence and prognosis of stroke in PD, AD and other neurodegenerative diseases. Therefore, the research results support the medical application of the panaxadiol saponin optimal composition in preventing and treating various neurodegenerative diseases and apoplexy and malignant prognosis thereof from the perspective of neurovascular units.
Example 10. In conditions of low or absent mitochondrial Complex enzyme I function, diseased vascular endothelial cells release inflammatory factors and drive astrocytes to convert to A1 toxic cell types, leading to a broad and deep progression of the disease network; the optimal composition induces the formation and development of PD disease networks involving endothelial cells, astrocytes and neurons starting from protecting endothelial cells against reduced or absent complex enzyme I function.
As described above, although it is known that the function and structural degeneration or damage of neurovascular unit (NVU) is closely related to neurodegenerative diseases and diseases such as stroke and sequelae thereof, it is not known how different member cells interact in NVU during occurrence and development of related diseases, such as who first attacks, lesions and affects other member cells, etc., and thus it is not possible to suppress diseases in early stages or sprouting states by precisely targeting upstream pathological events or the most fragile member cells, nor is it possible to accurately understand the partnership and action mechanism of a preferred composition, especially the partnership and cooperation of functional units thereof, at the NVU level.
The results of the study in example 9 demonstrate that brain microvascular endothelial cells are the most susceptible or vulnerable cells in NVU to loss of function of complex enzyme I by ROT, and that PDSC1 protects cells of each member of NVU. On this basis, further explore whether pathological products such as inflammatory factors are released by pathological endothelial cells, and further make peripheral astrocytes and neurons vulnerable to the decrease or deletion of the original complex enzyme I? But damaged astrocytes can further exacerbate nerve damage? Thus, PDSC1 protects individual member cells in the neurovascular unit against ROT toxicity, thus preventing the formation and deep progression of the ROT-induced PD disease network? For this purpose, the following studies were conducted in order to verify these ideas.
Example 10-1. Whether a culture medium for ROT-induced diseased endothelial cells makes astrocytes and neurons vulnerable to ROT?
The conditioned medium itself (ECM-ROT 50 nM) from which endothelial cells were cultured for 48h at low concentration of ROT (100 nM toxic to endothelial cells only in the neural unit) was compared to its addition of 100nM ROT (ECM-rot+rot) to induce apoptosis of Astrocytes (ASC) and dopamine neuronal PC12 cells. Meanwhile, parallel studies were performed on conditioned medium of endothelial cells protected by PDSC1, and corresponding control groups including normal control group (ROT, ECM-ROT) and normal culture conditioned medium group (ROT, ECM-ROT+0) were set. Conditioned medium was prepared by conventional methods: the culture medium of HBMEC for 48h was filtered to remove residual ROT using a 3kDA ultrafiltration tube, and the membrane-enriched proteins, including inflammatory factors, were routinely eluted and concentrated prior to dilution to obtain a specific conditioned medium (ECM-ROT) for culturing astrocytes and neurons for 48h. The apoptosis rate of each group of cells was determined using the conventional method annexin v fluorescent double-stain apoptosis kit.
The experimental results are shown in table 40: corresponding to the pre-cell viability results, low dose rotenone (100 nM) only induced 14.00% Astrocyte (ASC) apoptosis, but the lesion HBMEC conditioned medium induced by ROT (100 nM) was able to induce 27.00% astrocyte apoptosis, whereas the treatment of HBMEC conditioned medium with a non-damaging concentration of ROT (25 nM) did not induce astrocyte apoptosis. The apoptosis rate of PC12 cells is obviously improved by the two conditional culture media, which are 19.00 percent and 38.00 percent respectively. In particular, when 100nM ROT was added to the conditioned medium, the astrocyte apoptosis rate reached 56.00%, which was significantly higher than that of the control group (p < 0.001) and the low concentration conditioned medium group (ROT-ECM) to which only 100nM ROT was administered. The same results were seen in PC12 cells, with 100nM ROT itself causing only 19.00% apoptosis in PC12 cells, whereas conditioned medium plus 100nM ROT damage resulted in apoptosis rates as high as 71.87% significantly higher than that caused by ROT alone (p < 0.001) and ROT low concentration conditioned medium group (ROT-ECM). The apoptosis rate of astrocytes and neurons induced by the PDSC 1-protected endothelial cell conditioned medium itself (PDSC1+ROT-ECM) and its plus 100nM ROT (PDSC1+ROT-ECM+ROT) was significantly lower than that of the ECM-ROT conditioned medium and its plus ROT treated group.
Table 40 cytotoxicity of vascular endothelial cell-damaging conditioned Medium on astrocytes and neurons and protective action of PDSC1
Figure BDA0004007353830000441
ROT: a rotenone model group; ROT-ECM: rotenone damaging brain microvascular endothelial cell conditioned medium; *** p<0.001 (vs. normal control group); ### p<0.001 (vs. ROT group); && p<0.01 (vs. ROT-ECM group); @@@ p<0.001 (vs. ROT+ROT-ECM group); n=5.
The above results demonstrate that the release of toxic substances (or inflammatory factors) by ROT damaged HBMECs can significantly damage surrounding astrocytes and neurons, and in particular can greatly sensitize astrocytes and neurons to ROT toxic damage; PDSC1 protects endothelial cells against ROT toxicity and inhibits release of toxic substances by endothelial cells, thereby indirectly protecting astrocytes and neurons.
Example 10-2. Mitochondrial Complex enzyme I deficiency or loss of vascular endothelial cells the vulnerability of astrocytes and neurons to Complex enzyme I deficiency is increased by releasing inflammatory factors through the pro-inflammatory NF-kB pathway; PDSC1 inhibits complex I dysfunction or loss of activation of NF-kB pathways and inflammatory factor release, thus maintaining neurovascular units exposed to PD risk factors from morbidity.
It is known that inflammatory pathway activation usually occurs in diseased vascular endothelial cells, and thus the applicant believes that the diseased vascular endothelial cells caused by ROT become vulnerable to the deficiency or absence of mitochondrial complex enzyme I function by releasing inflammatory factors from peripheral astrocytes and neurons. To verify this assumption, a comparative study was first made of whether 100nM ROT selectively activates endothelial cell NF- κB inflammatory signaling pathways of neurovascular units (NVU) and releases a large number of pro-inflammatory factors into the culture medium, while 25 μM PDSC1 was observed to be likely to be acting against the ROT-induced inflammatory response.
Each member cell of NVU was cultured in a conventional manner, including mouse microvascular endothelial cells (bEnd.3), primary mouse Astrocytes (ASC) and microglial cells (MGA), and dopamine neuron PC12 cells, and when the cell abundance reached 70-80% were exposed to 100nM ROT 48h, respectively, the cells were collected for measuring the mRNA levels of intracellular NF- κB inflammatory signaling pathway activation indicator phosphorylating p65 (p-NF- κ B p 65) and downstream major inflammatory factors including TNF- α, IL-1α, IL-1β, IL-6, iNOS, ICAM, VCAM and RELA, and the protein levels of inflammatory factors in the activated member cell culture medium. The level of p-p65 was analyzed by Western Blot, mRNA and protein levels of inflammatory factors were analyzed by qPCR and ELISA kits, respectively, and NO levels were analyzed by kit methods.
The results of the study showed that a ROT selectivity of 100nM increases the p-NF- κ B p65 level of each member cell (FIG. 26), accompanied by a significant increase in the mRNA levels of intracellular TNF- α, IL-1β, IL-6, iNOS, ICAM, VCAM and RELA (Table 41) and a significant increase in the TNF- α, IL-1β, IL-6 and NO levels in the medium (Table 42); PDSC1 almost completely antagonizes the mRNA levels of ROT up-regulated endothelial inflammatory factors and adhesion molecules and releases inflammatory factors into the culture medium. However, PDSC1 slightly increases the level of iNOS and its product NO in normal endothelial cells but causes cellular inflammatory response, thus contributing to the alleviation of the state of vascular tone.
TABLE 41PDSC1 inhibits the mRNA levels of Rotenone (ROT) up-regulating pro-inflammatory factors and adhesion molecules of endothelial cells
Figure BDA0004007353830000451
CON: normal control group; PDSC1 panaxadiol saponin composition 1; ROT: rotenone; *** p<0.001(vs.CON); ### p<0.001(vs.ROT);n=5。
the above results demonstrate that low concentrations of rotenone (ROT, 100nM, inhibiting mitochondrial complex enzyme I thus results in insufficient or even absent function of this enzyme) selectively activate the vascular endothelial cell pro-inflammatory NF- κb pathway of neurovascular units (NVU), thus resulting in endothelial cell release of adhesion molecules and pro-inflammatory factors; the PDSC1 can almost completely maintain the vascular endothelial cell pro-inflammatory NF- κB pathway activity steady state with insufficient mitochondrial complex enzyme I function, so that the PDSC can prevent the PDSC from releasing pro-inflammatory factors to other peripheral brain cells. It should also be noted that PDSC1 upregulates NO release levels to about 30% for both normal and deficient endothelial cells of mitochondrial complex I, thus contributing to the prevention and relief of vascular tone and thus to the improvement of oxygen and nutrient supply to the hub.
Table 42PDSC1 inhibits Rotenone (ROT) from upregulating pro-inflammatory factors released by endothelial cells
Figure BDA0004007353830000452
CON: normal control group; PDSC1 panaxadiol saponin composition 1; ROT: rotenone; *** p<0.001(vs.CON); ### p<0.001(vs.ROT);n=5。
based on this, it was further determined whether the inflammatory factor released by the endothelial cells is an important cause of the damage to the otherwise existing deficiency of the function of the harmless mitochondrial complex enzyme I by other member cells in NVU caused by the pathological endothelial cell conditioned medium. For this purpose, the inflammatory environment of the pathological endothelial cell conditioned medium was reconstituted with commercial inflammatory factors and observed for the reproducibility of the pathological effects of the toxic conditioned medium of example 10-1 on astrocytes and neurons, and then the results of the study were reflected by the selective inhibition of endothelial cell p-65 mediated inflammatory activation with small interfering RNAs (siRNAs).
The proinflammatory factor levels in the media of the ROT-treated endothelial cells shown in Table 42 were measured by reconstituting 100nM ROT-treated vascular endothelial cells containing 5 μg/ml TNF- α, 15 μg/ml IL-1β and 10 μg/ml IL-6 for 48 hours of conditioned media with commercial inflammatory factors and observing their effects on induction of astrocyte and dopamine neuron PC12 apoptosis alone or in combination with 100nM ROT.
As shown in Table 43, the conditioned medium reconstituted with pro-inflammatory factors completely reproduced the pathological effects of the medium of 100nM ROT for 48 hours in vascular endothelial cells, as evidenced by the fact that the conditioned medium reconstituted with pathogenic inflammatory factors itself reduced to some extent the cell viability of astrocytes and PC12 cells, while greatly increasing the toxicity of ROT to both cells.
TABLE 43 proinflammatory factors of rotenone damaging conditioned Medium can completely mimic the toxicity of conditioned Medium to astrocytes
Figure BDA0004007353830000453
Figure BDA0004007353830000461
CON: normal control group; ROT: rotenone; *** p<0.001(vs.CON); ### p<0.001(vs.ROT 100nM);n=5。
as shown in table 44, conditioned medium of p 65-inhibited endothelial cells treated with 100nM ROT for 48h did not increase the apoptosis rate of Astrocytes (ASC) and PC12 cells nor the apoptosis rate of these two cells induced by low concentrations of ROT.
Conclusion and discussion
The above results, combined with the results of example 10-1, demonstrate that a 100nM ROT, which is toxic to endothelial cells alone, selectively activates endothelial pro-inflammatory NF-kB pathways in NVU and causes them to release adhesion molecules (including ICAM-1, VCAM-1) and inflammatory factors (including TNF- α, IL-1β, IL-6), where the combined action of TNF- α, IL-1β and IL-6 is sufficient to render astrocytes and dopamine neurons extremely fragile/vulnerable to otherwise non-pathogenic mitochondrial complex enzyme I dysfunction.
In addition, leukocytes in the blood interact with endothelial cells and undergo inflammatory infiltration through the blood brain barrier, and further exacerbate the inflammatory response in the brain and various neurodegenerative pathological processes including PD and AD. This process is driven by the adhesion molecules on the surface of the endothelial cells of the cerebral vessels (ICAM-1, VCAM-1, etc.). Therefore, surface adhesion molecules upregulated by endothelial cells with insufficient mitochondrial complex enzyme I function (PD risk factor) play a role in the entry of wolves (leukocytes) into the compartment (brain), further complicating the pathological mechanisms of PD.
The ginsenoside-containing PDSC1 can protect vascular endothelial cells with insufficient mitochondrial complex enzyme I function, inhibit NF- κB pathway, adhesion factors and inflammatory factor release, so that the cells of each member of the neurovascular unit exposed in PD risk factors can be maintained without morbidity, and the function and structural homeostasis of the neurovascular unit can be maintained. Since small RNAs (miRNAs) regulate the expression of these adhesion molecules by two different major mechanisms, namely by modulating the pro-inflammatory NF- κb pathway (controlling its transcription) and directly targeting them, the effect of PDSC1 to completely inhibit the upregulation of adhesion molecules by endothelial cells with insufficient mitochondrial complex enzyme I function is also indicative of the effect of PDSC1 to maintain the functional homeostasis of miRNAs.
TABLE 44 Selective inhibition of endothelial cells p65 with siRNA eliminates toxicity of Rotenone (ROT) treated endothelial cell conditioned medium to astrocytes and neurons
Figure BDA0004007353830000462
CON: normal control group; ROT: rotenone; ROT-ECM: rotenone (ROT) intervenes in the brain microvascular endothelial cell conditioned medium; ROT-sip65-ECM: siRNAp65 in combination with Rotenone (ROT) intervenes in brain microvascular endothelial cell conditioned medium; *** p<0.001(vs.CON); ### p<0.001(vs.ROT 100nM); &&& p<0.001(vs.ROT-CEM+ROT);n=5。
example 10-3. Release of inflammatory factors by diseased endothelial cells neurotoxic A1 astrocyte type.
Astrocytes are in an A2 state and have neuroprotection, while A1-state cells, which are pathological cells of astrocytes, lose physiological functions such as neuroprotection and maintenance of microenvironment homeostasis such as neurotransmitters in normal states, and release pro-inflammatory factors to cause neuroinflammation, and the increase of A1 cell numbers is closely related to PD and AD, so that A1 cells are also called neurotoxic A1 cells. Inflammatory factors released by microglia are known to induce A2 to be converted into A1 state, so that it is reasonable to consider that inflammatory factors released by diseased endothelial cells also induce A1 to form, thereby further deepening and widening the PD pathological network; similarly, PDSC1 protects endothelial cells with insufficient mitochondrial complex I function and thus avoids the A1 formation and its malignant pathological consequences induced by such endothelial cells. To verify this hypothesis, the effect of 100nM of ROT-induced pathological endothelial cell conditioned medium (ROT-ECM) treatment of astrocytes for 48h to induce A1 cell formation was observed, while the normal culture Control (CON), 100nM of ROT-treated control (ROT) and normal endothelial cell conditioned medium control (ECM) were set. The detection indexes comprise specific gene transcription products H2-D1, H2-T23 and A2 specific products sphk1 and B3gnt5 of A1 cells, astrocyte specific protein GFAP and A1 cell specific protein C3D. qPCR was used to determine the level of the selected gene transcript.
The results of the study (Table 45) show that the ROT-induced diseased endothelial cell conditioned medium (ROT-ECM) significantly increased the mRNA levels of H2-D1 and H2-T23 of astrocytes, while the low concentration of ROT (100 nM) fraction and slightly increased the mRNA levels of H2-D1 characteristic of A1, while significantly increasing the mRNA levels of sphk1 and B3gnt5 characteristic of A2. Consistent with the effect of PDSC1 protection of endothelial cells against ROT toxicity, the effect of PDSC 1-protected ROT-treated endothelial cell conditioned medium (PDSC 1-ROT-ECM) was consistent with the effect of low concentration of ROT (ROT), slightly increasing the A1 phenotype while increasing the A2 phenotype. Research results prove that the pathological endothelial cell culture medium can convert normal astrocytes into an A1 state, the low-concentration ROT without cytotoxicity promotes the conversion of the normal astrocytes into an A2 state, and the PDSC1 can inhibit the endothelial cells with insufficient mitochondrial complex enzyme I function from releasing substances for promoting the A1 phenotype and simultaneously increase the release of substances for promoting the A2 phenotype.
Table 45ROT induces the release of inflammatory factors by diseased endothelial cells to induce the neurotoxic astrocyte A1 phenotype
Figure BDA0004007353830000471
CON: normal control group; ROT: rotenone; ROT-ECM: the rotenone interferes with the brain microvascular endothelial cell conditioned medium; * p<0.05, *** p<0.001(vs.CON); ### p<0.001(vs.ROT-ECM);n=5。
Conclusion and discussion:
in combination with the results of the studies of examples 10-1 to 10-3, the applicant found that, under the conditions of exposure to the deficiency of mitochondrial complex I function, only pro-inflammatory NF- κB pathways of endothelial cells are activated and secrete inflammatory factors such as IL-1β, IL-6 and TNF- α by each member of the neurovascular unit including microglia, and that the combined action of these three pro-inflammatory factors is sufficient to induce the astrocyte A1 phenotype, the ginsenoside-optimized composition PDSC1 can protect endothelial cells against the impact of the deficiency of complex I function, thereby greatly slowing down the activation of pro-inflammatory NF- κB pathways and the secretion of pro-inflammatory factors and thus preventing A1 cell formation.
The combined actions of IL-1 beta, TNF-alpha and C1q derived from pathologically activated microglia are known to induce the astrocyte A1 phenotype and are believed to be important causative mechanisms of neurodegenerative diseases including AD and PD. Our research findings indicate that diseased endothelial cells are also the etiology of A1 formation, and that diseased endothelial cells may be the sole A1 cell cause in early PD (the primary cause of mitochondrial complex I dysfunction), thus updating understanding of the mechanisms underlying the development and progression of PD and other neurodegenerative diseases. In addition, brain microvascular destruction is a common feature of various neurodegenerative diseases including AD and PD, and the pathological role of brain microvascular destruction in neurodegenerative diseases has been understood in the past mainly from the standpoint of energy, nutrient and oxygen supply and blood brain barrier. Our findings further reveal the mechanism by which inflammatory endothelial cells initiate and promote neurodegenerative diseases by inducing A1 cells and adhesion molecules to the compartment (brain) by wolves (peripheral leukocytes). Our research findings therefore indicate that protecting cerebrovascular endothelial cells is a promising new strategy for the prevention and treatment of neurodegenerative diseases including AD and PD and cerebral apoplexy sequelae, and that PDSC1 and other advantageous compositions can implement this therapeutic strategy.
Example 10-4. Diseased endothelial cells directly poison nerve cells and increase their vulnerability to deficiency in Complex enzyme I function by inducing A1 formation and impairing Complex enzyme I deficiency to induce A2.
Based on the results of the study of example 10-3, the levels of pro-inflammatory factors (including TNF-. Alpha., IL-1β and IL-6) and anti-inflammatory factors (including IL-4 and IL-10) were further determined by 100nM of the conditioned medium (ROT-ECM) of ROT-treated endothelial cells, while the corresponding control group was set up to include normal culture astrocytes (CON), 100nM of the ROT-treated astrocytes (ROT, actually A2), and conditioned medium (ECM) of normal endothelial cells.
As shown in Table 46, the conditioned medium (ROT-ECM) of ROT treated endothelial cells induced A1 cells to release the pro-inflammatory factors TNF- α, IL-1β and IL-6 to the medium at levels significantly higher than those of the other control groups, but the anti-inflammatory factors IL-4 and IL-10 were significantly lower than those of the other control groups, while the ability of A2 cells to release the anti-inflammatory factors tended to increase. This suggests that A1 cells lose the ability of normal astrocytes to release anti-inflammatory factors, while releasing large amounts of pro-inflammatory factors. Importantly, the endothelial cell conditioned medium protected by PDSC1 (PDSC 1-ROT-ECM) did not significantly up-regulate the levels of pro-inflammatory factors secreted by astrocytes, but rather had a tendency to increase anti-inflammatory factor secretion. This effect of PDSC1 is consistent with its effect on conditioned medium (PDSC 1-ROT-ECM) in inhibiting A1 phenotype formation and tending to promote A2 formation (Table 45).
TABLE 46 characterization of inflammatory factor release by ROT-induced A2 cells and ROT-treated endothelial cell culture medium-induced A1 cells
Figure BDA0004007353830000472
Figure BDA0004007353830000481
CON: normal control group; ROT: rotenone; ROT-ECM: rotenone is used for treating brain microvascular endothelial cell conditioned medium; * p<0.05, *** p<0.001(vs.CON); ### p<0.001 (vs. ROT-ECM induced A1); n=5.
Next, the effect of A1 cell conditioned medium (A1 CM) and A2 cell conditioned medium (A2 CM) on themselves to induce apoptosis of PC12 cells, and their effect on the induction of apoptosis of PC12 cells in combination with 100nM Rot, A1CM-Rot and A2CM-Rot, respectively, were examined, while the respective control groups including normal Control (CON), 100nM Rot control (ROT) were set. The results of the study are shown in Table 47, and the apoptosis rate of PC12 cells induced by the combination of the A2 cell culture medium and the ROT is obviously lower than that induced by the ROT of 100nM, which indicates that the A2 cells can release neuroprotective substances; the apoptosis rate (44.03%) induced by the A1 cell conditioned medium per se is greatly higher than the apoptosis rate (18.26%) induced by 100nM ROT, and the apoptosis rate (69.43%) of PC12 induced by 100nM ROT is remarkably improved, which indicates that inflammatory factors released by A1 cells are sufficient to induce peripheral nerve cells to apoptosis, and nerve cells with insufficient mitochondrial complex enzyme I function are extremely vulnerable to toxicity of A1. The rate of neuronal apoptosis induced by the conditioned medium of the ROT combined PDSC1 treatment of astrocytes (PDSC 1-ACM) (22.36%) was very significantly lower than that induced by the A1 cell conditioned medium (44.03%) and the ROT combined A1 cell medium (69.43%).
TABLE 47 influence of A2 and A1 cell Condition Medium on neuronal apoptosis in combination with ROT, respectively
Figure BDA0004007353830000482
CON: normal control group; ROT: rotenone; a1CM: a1 astrocyte conditioned medium; a2CM: a2 astrocyte conditioned medium; PDSC1-ACM: PDSC1 treatment of astrocyte conditioned medium; ** p<0.01, *** p<0.001(vs.CON); # p<0.05, ### p<0.001(vs.ROT); @@@ p<0.001(vs.Rot+A1CM); &&& p<0.001(vs.A1CM);n=5。
taken together, the above studies demonstrate that inflammatory endothelial cells can damage neurons by a quadruple mechanism under conditions where neurovascular units are exposed to mitochondrial complex enzyme dysfunction, including induction of impaired astrocyte physiological function (including maintenance of central homeostasis by clearance of extracellular excess Glu and cellular metabolites such as α -synuclein), reduction of neuroprotective A2 formation, increase of neurotoxic phenotype A1 formation to directly poison neurons, and increase of vulnerability of complex enzyme I dysfunction to neurons. PDSC1 breaks the mechanism of these quadruple injury neurons by protecting endothelial cells against the impact of insufficient mitochondrial complex enzyme function.
Example 10-5. Normal endothelial cells increase the ability of astrocytes to phagocytize pathological products. A1 cells induced by endothelial cells with insufficient complex enzyme I function lose phagocytic ability, and A2 cells induced by complex enzyme I function acquire stronger phagocytic ability.
Based on the findings of example 10-4, the functions of astrocytes in basal state and phagocytosis in A1 and A2 states to clear pathological products were further compared. The ability of astrocytes to phagocytose pathological products was determined using fluorescent microsphere phagocytosis experiments, with higher intracellular fluorescence intensity indicating greater phagocytic ability. The test group included: normal control astrocytes (CON) in basal state, 100nM ROT treated astrocytes (A2 state (ROT/A2), normal endothelial conditioned medium treated astrocytes (ECM) and ROT-induced complex enzyme I-deficient endothelial conditioned medium treated astrocytes (A1 state (ROT-ECM/A1).
The results of the study are shown in Table 48, in which the ability of 100nM of the ROT-induced astrocytes in the A2 state to phagocytose fluorescent microspheres is significantly higher than that of cells in the basal state (CON), while the phagocytic ability of ROT-induced complex enzyme I function-deficient endothelial cell condition-induced A1 cells (ROT-ECM/A1) is very significantly lower than that of cells in the basal state; surprisingly, normal endothelial cell conditioned medium (ECM) also significantly increases phagocytic capacity of astrocytes, indicating that the active substance released by normal endothelial cells increases phagocytic capacity of astrocytes; the effect of PDSC1 plus ROT treatment on endothelial cell conditioned medium (PDSC 1-ROT-ECM) also significantly increased astrocyte phagocytic capacity compared to the normal control group, consistent with the effect of this conditioned medium on A2 cell formation.
Table 48 Effect of endothelial cell Condition Medium and A2, A1 phenotype on astrocyte phagocytic Capacity
Figure BDA0004007353830000491
CON: normal control group; ROT: rotenone; ECM, normal brain microvascular endothelial cell conditioned medium; ROT-ECM: the rotenone interferes with the brain microvascular endothelial cell conditioned medium; PDSC1-ROT-ECM: PDSC1 plus ROT treatment of endothelial cell conditioned medium; * p<0.05, *** p<0.001(vs.CON); ### p<0.001 (vs. ROT-ECM induced A1); n=5.
It can be seen that the results of the studies herein, in combination with the results of examples 10-1 to 10-4, demonstrate that endothelial cells with insufficient mitochondrial complex I function not only lose their function of enhancing astrocyte phagocytic capacity by releasing active substances, but also greatly impair astrocyte phagocytic capacity by releasing inflammatory factors, resulting in conversion of astrocytes into the A1 phenotype; PDSC1 not only inhibits the formation of A1 by the deficient mitochondrial complex enzyme I function of endothelial cells releasing inflammatory factors, but also maintains its function of releasing active substances that promote the A2 phenotype. Clearly, under global conditions, a decrease in astrocyte phagocytic capacity may lead to abnormal deposition of gamma-synuclein (gamma-syn), thus further deepening the PD disease network; PDSC1 can prevent the formation and development of PD disease networks by protecting vascular endothelial cells. In examples 10-6, we will verify this reasoning.
Summary and discussion of the results of the study of examples 10-1 to 10-5:
comprehensive examples 10-1 to 10-5 studies found that the following conceptual conclusions can be drawn: the deficiency of the function of the mitochondrial complex enzyme I of the PD risk factors can selectively or preferentially activate the NF- κB pro-inflammatory pathway of the vascular endothelial cells and release inflammatory factors, thereby weakening the beneficial effect of the endothelial cells on astrocytes; at the same time, A1 formation is induced, thereby losing astrocyte protective function and obtaining the neurotoxicity of A1 cells, thus greatly widening the network of PD diseases which deepen inflammatory endothelial cell initiation, including endothelial cell injury and loss, brain microvascular injury, blood brain barrier destruction, astrocyte loss, neuroinflammation, abnormal α -syn aggregation and neuronal injury. In addition, inflammatory endothelial cells up-regulate surface adhesion molecules play a role in the entry of wolves (white blood cells) into the chamber (brain), thus further complicating the network of PD disease. It can be seen that protecting the cells of each member of the neurovascular unit, particularly the fragile endothelial cells therein, is critical for the prevention and treatment of PD and other neurodegenerative diseases. PDSC1 can prevent and treat PD important risk factors (mitochondrial complex enzyme I insufficiency) by protecting individual member cells of neurovascular units to trigger the formation and development of PD disease networks; meanwhile, the PDSC1 can also improve or maintain the physiological functions of vascular endothelial cells, including releasing more NO to improve the cerebral vascular tension state and cerebral blood circulation disorder caused by the cerebral vascular tension state, promoting the conversion of astrocytes to neuroprotective phenotype A2 cells and releasing neuroprotective active substances, and avoiding the formation of A1 cells, thereby improving the functions of neurovascular units and the coping capability of mitochondrial complex enzyme I functional insufficiency.
From the results of the studies in the prior examples, it was predicted that in the ROT-induced PD rat brain, the vicious circle of mutual injury between the individual member cells of the neurovascular unit initiated by inflammatory endothelial cells will eventually manifest as blood brain barrier disruption, microvascular and endothelial cell injury and peripheral astrocyte loss, and PDSC1 can protect the blood brain barrier, microvascular, endothelial cells and their peripheral astrocytes.
Examples 10-6. Rotenone (ROT) -induced rat PD model also presents a chain pathological response of inflammatory endothelial cells to neurovascular units, PDSC1 can prevent ROT-activated NF- κb and subsequent cytopathic and damaging members of neurovascular units; the preferred compositions start from protecting endothelial cells against the formation and development of PD disease networks involving endothelial cells, astrocytes and neurons.
Here, the studies of examples 10-1 to 10-5 were validated using the ROT-induced rat PD model and found to lead to conceptual conclusions (inflammatory endothelial cells initiate the effects of malignant linkage reactions between member cells of neurovascular units on the formation and development of the PD disease network; PDSC1 can control the formation and development of the PD disease network by protecting vascular endothelial cells from insufficient function of the essential risk factor mitochondrial complex enzyme I for PD). To reveal the substantial differences in pharmacological effects of PDSC1 and dopamine replacement therapy L-DOPA, the pharmacological effects of certain indicators of PDSC1 (40 mg/kg/day, the effective dose found previously) and L-DOPA (metaba, equivalent to 7 times the initial dose of PD patients) were also studied in comparison. To determine whether NF- κ B p65 activation (marker p-p 65), A1 cell presence (marker C3D), microglial activation (marker iba 1), abnormal deposition of α -syn (soluble and insoluble non-phosphorylated and phosphorylated forms), α -syn phosphorylation marks pathological protein transmission in the brain), endothelial cell and dopamine nerve (marker TH) death occurred and their order in the PD rat brain, the observation points before the ROT (D0) and the next day after the ROT (D2), 10 TH day (D10) and 15 TH day (D15) were set, the brain areas including striatum and substantia nigra compact were observed, and the examination indexes were set using Western Blot (WB, analysis of striatum only) and immunohistochemical assay. In addition, blood brain barrier and brain microvascular damage were also observed for the 15 th day specimens. Delivering fluorescent marker proteins to the cerebrovascular network by cardiac perfusion to detect the damage of the blood brain barrier; EBA and GFAP are used as markers of endothelial cells and astrocytes, respectively, and fluorescent immunostaining and double staining methods are used to observe the morphology of the microvessels and their relationship with surrounding astrocytes.
WB analysis results showed (fig. 27, table 49 and table 50) that NF- κb was significantly activated and began to appear in the striatum on day 2 (D2) with no significant changes in other indices in both striatum and substantia nigra compacta compared to before ROT (D0); on day 10 (D10), NF- κb activation (p-p 65 up-peak) and A1 cell numbers peaked (C3D up-peak), microglial activation (iba 1 up-peak), soluble and insoluble non-phosphorylated and phosphorylated α -synuclein levels were elevated to peak. Consistently, immunohistochemical results (fig. 28) showed that p65 staining of striatum appearance presented vessels starting on day 2; the astrocytes appear in the cell body and the protrusion are separated, especially the vascular structure formed by the astrocyte end plates is greatly disappeared compared with the normal animal (D0) with incomplete structure and surrounding GFAP positive fibers, and the pathological change of the astrocytes is aggravated along with the prolonged time; at this time, no morphological change of microglial cells was seen, nor was iNOS expressed by cerebral vessels. By day 10, microglial appearance of amoeba status indicated that it was activated (a in fig. 27), inflammatory vascular endothelial cell expression iNOS staining (C in fig. 28), dorsal striatal outer side (this brain region mainly received projections from sensory motor cortex limb representative region, with significant effects in performance of normal motor function, learning of motor skills and habit formation) TH level significantly reduced (D in fig. 28), but no significant changes in substantia nigra compact part; on day 15 TH was further reduced in the striatum and the substantia nigra pars compacta dopamine nerve fiber TH was almost completely lost. It can be seen that, in the ROT-induced PD rat model, vascular endothelial cell NF- κ B p65 activation, A1 cell appearance, pathological α -syn transmission in striatum and substantia nigra and its abnormal aggregation, and the occurrence sequence of dopamine nerve injury were highly consistent with those found in the cell model. It is also specifically noted that microglial activation occurs after endothelial NF- κb activation and A1 cell appearance, and this sequence is also consistent with the 100nM activation of only endothelial NF- κb without affecting microglial and other cells in the neurovascular unit, again suggesting that inflammatory factors released by endothelial cells during the development of ROT-induced PD generation are the first driving forces for astrocyte conversion to A1 phenotype, and that subsequent pathologically activated microglial cells may also be added to the lines promoting A1 conversion, as reported in the literature.
TABLE 49 effects of ROT on the development and progression of neurovascular units and PDSC1 in the development of PD rat models
Figure BDA0004007353830000501
* p<0.05, ** p<0.01, *** p<0.001 (vs. normal control group); ### p<0.001 (vs. 15 th day ROT model group); n=3.
TABLE 50 action of ROT-induced PD on alpha-synucleinopathy Process and PDSC1 in the Generation and Generation of rat models
Figure BDA0004007353830000502
** p<0.01, *** p<0.001 (vs. normal control group); ### p<0.001 (vs. 15 th day ROT model group); n=3.
Consistent with the pharmacological actions obtained in the cell model, PDSC1 can inhibit the proliferation of ROT chronic treatment induced endothelial NF- κb activation, astrocyte transformation to A1 phenotype, microglial activation, toxic α -syn transmission and abnormal aggregation, and protect striatum and nigral dopamine neurological pathways and cerebral vessels. Specifically, PDSC1 was administered 45 minutes prior to each ROT administration and could completely prevent ROT-induced NF- κb activation indicated by elevated p-p65 and p65 levels, A1 cell appearance indicated by elevated C3d levels, and microglial activation indicated by elevated iba levels (fig. 27, tables 49 and 50); PDSC1 may also completely combat elevated levels of soluble phosphorylated and non-phospho α -syn in the striatum of ROT and significantly combat elevated levels of both non-soluble α -syn. Immunohistochemical results also showed that PDSC1 was almost completely resistant to ROT-induced striatal and substantia nigra compacta phosphorylated α -syn aggregation (B in fig. 27). It can be seen that PDSC1 is fully resistant to ROT-induced alpha-syn pathological processes, including toxic alpha-syn spread and abnormal aggregation. The research results are favorable for supporting the effect of the PDSC optimal composition on preventing and treating the neurodegenerative diseases from an important point of view based on the disease initiation and development processes of alpha-syn pathology involved in Parkinson's Disease (PD), dementia with lewy bodies (DLB), progressive Supranuclear Palsy (PSP), corticobasal degeneration (CBD) and Multiple System Atrophy (MSA). Consistent with the pharmacological actions described above, PDSC1 was almost completely resistant to ROT-induced loss of striatal dopamine nerve endings (D in fig. 28) and loss of substantia nigra dense dopamine nerve fibers (E in fig. 28), and L-DOPA did not exhibit significant neuroprotection, but its combination with PDSC1 did not negatively affect the efficacy of PDSC 1. The powerful protective effect is derived from the direct protective effect of the PDSC1 on the cells of each member of the neurovascular unit and the effect of preventing and treating the network occurrence and development of PD diseases caused by the damage of the cells of the neurovascular unit by inflammatory endothelial cells.
Fig. 29 and 30 further demonstrate the protective effect of PDSC1 on the vascular lesions and peripheral astrocytopathy in the striatum and substantia nigra brain areas of animals of the day 15 model control group. In the striatum of the brain region observed, the blood vessel structure in the brain of normal animals (naive) is complete, and the end plates of astrocytes are tightly adhered to the outer wall of the blood vessel to form a complete structure (A and B in FIG. 29); the rat with ROT model has a great loss of astrocytes around blood vessels, no close contact of astrocytes with the outer walls of blood vessels is found (the blood vessel structure is broken) (fig. 29A and 29B), which supports the conclusion that inflammatory endothelial cells aggravate the liability of astrocytes to ROT, L-DOPA has no obvious protective effect on astrocytes, and cannot protect the close connection between blood vessels and glia, PDSC1 and astrocytes in the combined group with L-DOPA are tightly connected with the outer walls of blood vessels, the structure of the glia-blood vessel unit is complete and almost consistent with that of the rat with normal group, which shows that PDSC1 almost completely resists the toxicity of the ROT-induced neurovascular unit, and the L-DOPA does not negatively affect the protective effect of PDSC1 when combined with L-DOPA, consistently, as shown in fig. 30A and B, the striatum of animals with ROT-induced PD model (A in fig. 30) and the dense part (SNc) and the ventral part (VTA) have great amounts of protein in the visible light, the fluorescent effect on the brain-DOSC has no obvious protective effect on the side part of the PDSC, and the contrast to the human brain-DOPA, and the side has no obvious protective effect on the side part of the PDSC has a strong toxic effect on the human-DOSC, but not affecting the human-DOPA, and the human-brain-DOPA has a very strong protective effect on the human-brain-vascular barrier, this strongly supports the medical use of effective PDSC in combination with L-DOPA for the treatment of PD, particularly middle and late stage PD, to compensate for the inability of L-DOPA to slow down disease progression.
Conclusion and discussion:
combining the results of the ROT-induced PD rat model study, the findings of examples 10-1 to 10-5 confirm the conceptual conclusions drawn, namely: the mitochondrial complex enzyme I deficiency can selectively or preferentially activate vascular endothelial cell NF- κB pro-inflammatory pathways and release inflammatory factors, thereby weakening the beneficial effects of endothelial cells on astrocytes, simultaneously inducing A1 type astrocytes to form, and the secondary microglial pathological activation can also be added into a line for promoting A1 type cell transformation, so that the physiological functions of the astrocytes are lost and the A1 cytotoxicity is obtained to widen together the PD disease network initiated by the endothelial cells, including endothelial cell injury and loss, brain microvascular injury, blood brain barrier damage, astrocyte loss, abnormal alpha-syn aggregation and blackish striatal dopamine pathway injury. Therefore, protecting the cerebrovascular endothelial cells can prevent the deletion of the complex enzyme I from starting the damage process to the neurovascular unit, and has important therapeutic value for preventing and treating PD. In addition, inflammatory endothelial cells up-regulate surface adhesion molecules before death of endothelial cells and astrocytes occurs, which can lead wolves (white blood cells) into the chamber (brain), further complicating the PD disease network. Compared with the disclosed drug targets for preventing and treating PD by using neurons, glia cells, various pathological events including inflammatory reaction, alpha-syn and the like, the drug targets for preventing and treating PD by using vascular endothelial cells are more scientific, feasible and efficient, and the feasibility and the efficiency of the drug targets are verified by the drug effects of the PDSC1 and other optimal compositions.
PDSC1 can prevent PD disease network initiation by protecting endothelial cells from low function of mitochondrial complex I, a risk factor for PD, thereby avoiding a subsequent series of linked pathological events including infiltration of peripheral immune cells across the blood brain barrier to brain tissue, A1-type cell formation, loss of astrocyte function, activation of microglial pathology, transmission and deposition of α -synuclein pathology, damage to the dopamine pathway in the substantia nigra, severe loss of vascular endothelial cells and peripheral astrocytes, and severe damage to the blood brain barrier. PDSC1 also increases the other physiological functions of endothelial cells and astrocytes on other members of the neurovascular unit, thus increasing the capacity of the entire neurovascular unit to cope with mitochondrial complex I insufficiency and correcting early pathological changes in PD and repairing the affected motor and neuropsychiatric disorders; PDSC1 also protects other member cells in the neurovascular unit directly against cytotoxicity caused by insufficient mitochondrial complex I, thus slowing or even cutting off the vicious circle between different member cells of NVU during the course of PD development, and thus slowing or even blocking the progression of the disease. The pharmacological actions reveal the pharmacological actions of the excellent-effect ginsenoside composition including the PDSC1 for preventing and treating the network formation and the deep progression of PD diseases by using a unique action mechanism system, so that the medical application of the excellent-effect ginsenoside composition for preventing and treating the PD from the root is shown in the treatment principle, and the stiffness of the long-term treatment and temporary treatment of the PD at home and abroad (the existing medicines all take the symptom relieving as the treatment target, have no medicine for delaying the disease progression and have no medicine for preventing the occurrence of the disease) can be broken. It is also specifically noted herein that brain microvascular lesions are also involved in the pathological processes of various atypical parkinsonism including multisystemic atrophy (MSA), progressive Supranuclear Palsy (PSP), corticobasal degeneration (CBD), frontotemporal lobar degeneration (FTLD) and dementia with lewy bodies (DLB), and these patients may exhibit a wide range of cognitive, neuropsychiatric, sleep, motor and autonomic neurological symptoms, and existing drugs including dopamine only slightly ameliorate motor symptoms therein. It can be seen that PDSC1 and other optimal compositions may also bring new promise to these patients.
It is also specifically noted herein that the results of the study strongly support the medical use of PDSC1 and other superior compositions for the prevention and treatment of other neurodegenerative and neurodegenerative disorders from a number of common pathological mechanisms. The effects of brain microvasculature on a variety of conditions including dementia and Alzheimer's disease, chorea (HD), progressive freezing syndrome (ALS), and Multiple Sclerosis (MS) are becoming increasingly recognized. For example, recent studies have shown that disruption of the Blood Brain Barrier (BBB) is an early biomarker of cognitive dysfunction in humans, including the early clinical stages of alzheimer's disease; various cerebrovascular risk factors (e.g., cholesterolemia, hypertension, etc.) and injury events (e.g., chronic low-perfusion, ischemic or hemorrhagic stroke, transient ischemic attacks, during stroke recovery) can cause neurodegeneration or severe injury by damaging neurovascular units and the blood brain barrier, thereby causing brain dysfunction or even disability; vascular lesions are also a significant cause of retinopathy. Amyloid deposition (assembly of polypeptides into amyloid fibers) is considered as a central pathological feature of another age-related disease, such as β -amyloid (aβ), phosphorylated tau protein (tau) and apolipoprotein E4 (APOE 4) in Alzheimer's Disease (AD), islet amyloid polypeptide (IAPP, amylin) in type II diabetes, alpha-synuclein (alpha-syn) and in Parkinson's Disease (PD), frontotemporal lobar degeneration (clinically manifested as frontotemporal dementia, FTLD), tau and TDP-43 proteins, and the like. Furthermore, one protein can accelerate the deposition of another protein, such as soluble IAPP, across the blood brain barrier, which when present with soluble aβ or α -syn accelerates the formation of amyloid, thus becoming an important cause of AD and PD in type II diabetics with far higher morbidity than in non-diabetic populations. Furthermore, a toxic protein is associated with a variety of diseases, such as α -syn pathology, and is also widely involved in the pathological processes of various atypical parkinsonism (including Progressive Supranuclear Palsy (PSP), corticobasal degeneration (CBD) and Multiple System Atrophy (MSA)), dementia with lewy bodies (dementia with lewy bodies and dementia with parkinson's disease, collectively referred to as dementia with lewy bodies (DLB), which is the second most common neurodegenerative and retinal degenerative diseases following AD, and the like, whereas ApoE4 is involved in the pathological processes of neurodegenerative diseases such as AD and frontotemporal lobar degeneration (FTLD) through aβ, tau and α -syn proteins and direct toxicity to nerves and cerebral blood vessels, and is also associated with traumatic brain injury and poor neurological prognosis following ischemia or hemorrhage. Neuroinflammation is another common pathological event in various neurodegenerative diseases including PD and AD. In this context it must be pointed out that astrocyte and microglial dysfunction and their interactions are closely related to the above-mentioned toxic protein deposition and neuroinflammation, and that pathologically activated microglial cells and A1 astrocytes not only debilitate toxic proteins but also release toxic substances such as pro-inflammatory factors to poison nerve cells and cerebral vessels. For a long time, the above common pathological events have been considered as respective relatively independent events, and thus a great deal of drug development such as an antibody against aβ, an antibody against α -syn, and the like has been performed for these pathological events, respectively, but most have been completed with failure in clinical trials. More and more studies have found that these pathological events are not independent processes, but rather are interrelated events in the disease network, which should be taken into account when developing a prophylactic treatment of neurodegenerative diseases. The results of the studies of examples 9 to 10 further indicate that disease risk factors such as mitochondrial complex I deficiency (for PD) are cytopathogenic to the most fragile members of the neurovascular unit (NVU), thereby initiating the formation and subsequent progression of the disease network towards depth and breadth, in which inflammatory vascular endothelial cells or/and pathologically activated microglia and their induction of A1 cell formation are early key nodes of the disease network, while A1 cells are further deepened by acquired toxicity and loss of astrocyte physiological function. The applicant believes, therefore, that enhancing the coping ability of the most fragile cells to the respective risk factors may counteract the onset of disease network formation, thus achieving the objective of delaying or even blocking the occurrence of the respective neurodegenerative disease; while protecting vascular endothelial cells and microglial cells, maintaining their functional homeostasis and avoiding the appearance and reversal of inflammatory states can protect the physiological functions of astrocytes and prevent and reduce A1 cell formation, thus achieving the objective of slowing or even blocking the progression of the disease network to depth and breadth.
Taken together, PDSC1 protects the pharmacological effects of NVU member cells, including the most fragile member cells, against PD risk factor mitochondrial complex I deficiency to induce cytopathy and prevent network formation and progression of PD disease, and directly promotes the formation of neuroprotective astrol A2 phenotype (release of neuroprotective active substances, greater ability to phagocytose toxic proteins) by enhancing endothelial cell release, suggesting that PDSC1 may enhance the ability of involved cells to cope with risk factors, reverse disease network embryonic form, slow or even block disease network extension and deep progression. It can be seen that PDSC1 and other superior compositions have important therapeutic value for the prevention and treatment of the various neurodegenerative diseases described above, retinopathy, treatment of traumatic brain injury and poor neurological prognosis following ischemia or hemorrhage.
Example 11. Efficacy study of the optimal composition PDSC1 to protect small albumin positive interneurons (PV-Ins).
To further verify the effects of PDSC1 in preventing neurodegenerative diseases, the broad neuroprotective effects of PDSC1 are further disclosed herein as subjects of inhibitory inter-neuronal small albumin inter-neurons (parkalbumin-expressing interneurons, PV-Ins, also known as fast-impulse inter-neurons) that are fragile and closely related to neurodegenerative diseases and mental behavioral disorders. PV-INs is an important inhibitory interneuron with GABA as transmitter, and is critical to maintaining the balance of the inhibitory neurotransmitter GABA and the excitatory transmitter glutamate (Glu), and also to regulate the release of other neurotransmitters, thus deeply involved in the physiological activities of the cerebral cortex and striatum. PV-INs dysfunction can directly lead to disruption of excitatory/inhibitory (E/I) balance and other related neurotransmitter disorders, leading to neuropsychiatric behavioral disorders including cognitive disorders and dyskinesias, etc., while Glu signaling persistence can lead to neurodegeneration including PV-INs, further deepening disease progression. PV-INs are particularly susceptible to environmental disturbances including extracellular high levels of Glu, mitochondrial membrane potential loss (ATP deficiency), oxidative stress and inflammation, and the like, and this vulnerability makes PV-INs dysfunction play an important role in many brain diseases including neurodegenerative diseases (such as AD and lewy body dementia) and cognitive decline associated with natural aging, mental disorders (such as schizophrenia and bipolar disorder), neurological disorders (such as epilepsy) and neurodevelopmental disorders (such as tourette syndrome, also known as tourette syndrome in children, childhood hyperactivity syndrome, also known as attention deficit disorder syndrome in children, autism). In the striatum, PV-INs exert a powerful inhibitory control effect, maintaining the firing rate of spiny neurons (SPNs) in the efferent nerves at low levels of physiological state. Consistently, the hyperexcitability of striatal Glu and the insufficient inhibitory activity of GABA are closely related to the motor symptoms of PD and the catabolism caused by L-DOPA treatment, and selective inhibition of PV-INs can also cause catabolism, so that the insufficient function of striatal PV-INs may also directly and indirectly participate in motor symptoms and mental disturbances of PD. Therefore, the study results of observing whether the PD rat striatum induced by the mitochondrial complex enzyme I inhibitor ROT has the function degradation of PV-INs and the protection effect of the PDSC1 on the PD rat striatum can further clarify the wide protection effect of the PDSC1 on neurovascular units and prevent and treat neurodegenerative diseases including various dementias, and also can disclose the medical application of the PDSC1 and other optimal compositions in preventing and treating mental disorder, nerve dysfunction diseases and nerve development disorder diseases and delaying the natural aging of the brain.
Striatal PV positive neuron density and positive fibers were observed by immunohistochemistry and immunofluorescence methods using the striatal samples of examples 10-6 to determine the function and survival of the PV-INs. The striatal segments (27 continuous slices from bregma 1.7 mm) were sectioned continuously (40 μm) and the brain pieces were collected as semi-quantitative. Briefly, for brain regions containing striatum, 3-piece collections (9 pieces per piece) were collected and stored per animal, 1 piece per piece was taken for analysis of the PV positive particle density, and the average of 3 pieces was used as data for one animal. Semi-quantitative analysis was performed on four regions of the Dorsally Lateral (DL), dorsally Medial (DM), ventral Lateral (VL) and Ventral Medial (VM) of the striatum. In particular, the PV (aSeed calcium binding proteins) are important functional proteins of PV-INs by binding to Ca 2+ Binding energy reduction of presynaptic Ca 2+ Level of Ca is thus strongly reduced 2+ Activated potassium ion conduction to exert its physiological function. Moreover, the level of expression of PV protein is positively correlated with the GABA level released by the PV cells, and thus its level of expression may also reveal the PV-INs region excitation/inhibition (E/I) equilibrium state. It can be seen that the level of PV immunostaining is positively correlated with the status of PV-INs function, and that the absence of staining does not necessarily mean that the neurons are lost, but indicates that their function is severely impaired or even lost.
As shown in Table 51, immunohistochemical staining showed that the PV-positive reduction occurred in all four areas of the striatum of the PD rat model group compared to normal animals, and in particular immunofluorescent staining showed almost complete loss of PV-positive fibers (FIG. 31); L-DOPA increased PV-positive particles in DL, DM, VL and VM compared to model groups, but no difference in selectivity was seen; the combination of PDSC1 and PDSC1 with L-DOPA can obviously increase PV positive particles in DL, DM, VL and VM ## p<0.01 And no significant differences were seen between the PV positive particles and fibers compared to the normal group. The research result shows that the striatum of the PD rat induced by ROT has extensive PV-INs function serious degradation, thereby revealing the pathological effect of the striatum PV-INs function degradation on the development of PD and the expression of neuropsychiatric state; PDSC can protect striatal GABA interneurons represented by PV-INs against the hazards of mitochondrial complex enzyme I deficiency/loss, further elucidating the broad protective role of PDSC1 on members of the neurovascular unit including PV-INs.
Table 51 ROT-induced reduction of PD rat striatum PV-positive GABA interneurons and protection of PDSC1
Figure BDA0004007353830000531
ROT: rotenone; L-DOPA: levodopa; PDSC1: panaxadiol saponin composition 1; ** p<0.01 (vs. normal control group); ## p<0.01 (vs. ROT model group); n=5.
Summary and discussion:
based on the physiological role of striatal PV-INs, the degeneration of PD striatal PV-INs function can directly participate in and even cause acanthose neuron (SPN) hyperexcitability to induce motor symptoms, and can cause or exacerbate striatal glutamate (Glu) hyperexcitability and other neurotransmitter level disorders to exacerbate motor symptoms and induce neuropsychiatric symptoms and accelerate disease progression. Therefore, the pharmacological effects of PDSC1 to protect striatal GABA interneurons, represented by PV neurons, against the hazards of mitochondrial complex I deficiency/loss predicts that PDSC1 and other optimal compositions can maintain and repair the effects of neuronal excitability and inhibitory balance in striatum and other brain areas involved in PD patients and breakthrough efficacy in preventing PD, including prevention of motor symptoms and central non-motor symptoms (including various psychotic symptoms and cognitive disorders) of PD, prevention of L-DOPA therapy-induced catabolism and neuropsychiatric disorders including delusions, hallucinations, and prevention of PD occurrence and development.
In particular, the ability of PDSC1 to increase the capacity of PV-INs to cope with the deficiency of mitochondrial complex I without onset reflects the broad protective effects of PDSC1 to protect PV-INs against other environmental interference factors and the medical use of PDSC1 to prevent brain diseases associated with PV-INs dysfunction, including: neurodegenerative diseases including Alzheimer's Disease (AD), lewy body dementia and frontotemporal dementia, cognitive decline associated with natural aging, psychogenic behavioral disorders (such as schizophrenia and bipolar disorder), epileptic formations and seizures, neurogenic developmental disorders (such as Creutzfeldt-Jakob disease, also known as Tourette's syndrome, childhood hyperactivity disorder, also known as childhood attention deficit disorder syndrome, autism), and stress psychological disorders and post-traumatic stress disorder.
It is also to be noted that, in terms of PV protein function (PV and Ca 2+ Binding energy reduction of presynaptic Ca 2+ Level of Ca is thus strongly reduced 2+ The effect of PDSC1 on the reduction of the level of anti-ROT-induced striatal PV positive staining and maintenance at levels close to normal animals reflects that PDSC1 can inhibit intracellular Ca by PV proteins 2+ Signal hyperactivity. Consistent with PV-INs dysfunction, nervesMeta-overactivity is a functional hallmark of Alzheimer's Disease (AD) in human and different mouse models and mediates memory and cognitive impairment. Moreover, recent studies have found that presynaptic calcium stores are determined to be a key factor in controlling AD-related neuronal overactivity and are also targets for disease treatment. This further supports the medical use of PDSC1 and other optimal compositions for the prevention and treatment of AD.
Example 12. Protection of cellular energy metabolism and redox homeostasis is an important mechanism by which a potent composition exerts its broad brain protective effects.
Rotenone (ROT) selectively inhibits mitochondrial respiratory chain complex enzyme I, thereby preventing electrons (mainly in the form of NADH) produced by the tricarboxylic acid (TCA) cycle from entering the oxidative respiratory chain, thereby inhibiting oxidative phosphorylation pathways, ultimately leading to mitochondrial dysfunction including reduced levels of Adenosine Triphosphate (ATP), redox imbalance and oxidative stress damage and subsequent inflammatory responses. This biochemical pathology is an important upstream mechanism of PD occurrence and development, and there is no drug capable of preventing and slowing down this upstream pathogenic event, so that even though PD patients have received various drug treatments including gold-labeled L-DOPA drugs, the disease continues to progress until the physical disability. From the studies of the previous examples, applicants have found that low concentrations of ROT can preferentially disrupt endothelial cell energy metabolism and redox homeostasis, whereas other member cells of the neurovascular unit, particularly astrocytes, have a stronger plasticity of energy metabolism and redox homeostasis, and therefore endothelial cells are most vulnerable to ROT; similarly, PDSC1 can increase cellular energy metabolism and redox plasticity, thereby protecting endothelial cells against ROT cytotoxicity. Accordingly, the following 3-aspect confirmatory study was conducted, and the study results not only demonstrate the biochemical essence of the endothelial cells on the vulnerability of mitochondrial complex enzyme I function deficiency and the protective effect of PDSC1, but also further reveal the commonality mechanism of the PDSC1 and other optimal compositions for preventing and treating neurodegenerative diseases and other mitochondrial dysfunction diseases.
Example 12-1 comparative study of the Effect of Low concentration of ROT on endothelial and astrocyte energy metabolism and redox homeostasis/ROT-preferential reduction of vascular endothelialCellular ATP and NAD + Level, disruption of its redox homeostasis and damage to mitochondria; astrocytes pass through to increase NAD + Level and NAD + the/NADH ratio is effective against mitochondrial complex enzyme I deficiency, and thus energy metabolism and mitochondrial function homeostasis in the condition of mitochondrial complex enzyme I deficiency can be maintained.
Brain Microvascular Endothelial Cells (BMECs) and Astrocytes (ASCs) were routinely cultured, and after 36h of ROT treatment at 100nM, the cell energy metabolism, oxidative stress and central carbon metabolic changes were examined, while the role of PDSC1 in metabolic regulation was analyzed. Various indices were determined by conventional methods. As shown in Table 52, after 36h of 100nM ROT treatment, the intracellular ATP level of BMEC was reduced by 50%, ADP was accumulated in large amounts, and the ATP/ADP ratio was reduced by 75% (p)<0.001). These data indicate that BMEC mitochondrial oxidative phosphorylation activity is disrupted. Accordingly, NADH (reduced coenzyme I) levels providing electron donors for oxidative phosphorylation pathways were increased to 325% (p)<0.001 While NAD + (nicotinamide adenine dinucleotide, also known as oxidized coenzyme I) tends to decrease, leading to NAD + the/NADH ratio was reduced to 21% (p)<0.001). In sharp contrast to endothelial cells, although mitochondrial oxidative phosphorylation of astrocytes was also significantly reduced, to a much lower extent than vascular endothelial cells, ATP levels and ATP/ADP ratios were reduced by 29% and 35%, respectively, significantly lower than 50% and 75% of endothelial cells. It can be seen that the capacity of the mitochondrial oxidative phosphorylation activity of astrocytes to resist deficiency or absence of complex enzyme I function is far greater than that of cerebrovascular endothelial cells. In particular, ROT treatment does not decrease but rather significantly increases NAD of astrocytes + Level (p)<0.01 While NADH levels are substantially unchanged such that NAD + the/NADH ratio was increased to 156%. It is to be noted here in particular that NAD + As a redox coenzyme, it is a center of energy metabolism and is important to maintain energy metabolism homeostasis. NAD in cytoplasm + Is a critical determinant of NADH and pyruvate flow rate to mitochondria and therefore glycolytic rate, or glycolytic activity is determined by NAD + The high score of the/NADH ratio. Due toNAD is required for TCA cycle and electron transporter channels, respectively + And NADH, thus an optimal NAD is required + NADH ratio to achieve efficient mitochondrial metabolism. Removal of cytoplasmic NAD + Preventing glycolysis and leading to cell death. NAD (NAD) + Deletions can lead to mitochondrial dysfunction, decreased energy production, and ROS accumulation, thereby creating high oxidative stress. Furthermore, NAD + Many key cellular functions other than energy metabolism, including DNA repair, chromatin remodeling and cellular senescence, can be directly or indirectly affected by the enzymes threshed, CD38 and poly (ADP ribose) synthase (PARP).
Table 52 endothelial energy metabolism is less plastic than astrocytes in response to mitochondrial complex enzyme I insufficiency
Figure BDA0004007353830000551
ROT: rotenone; ATP: adenosine triphosphate; ADP: adenosine diphosphate; ATP/ADP: ratio of adenosine triphosphate to adenosine diphosphate content; NAD (NAD) + : oxidized nicotinamide adenine dinucleotide; NADH: reduced nicotinamide adenine dinucleotide; NAD (NAD) + NADH: ratio of oxidized nicotinamide adenine dinucleotide to reduced nicotinamide adenine dinucleotide; *** p<0.001 (vs. normal control group); n=5.
Taken together, rotenone (ROT) induces endothelial cells NAD + Reduced level and NAD + The severe decrease in NADH ratio limits its response to reduced ATP production and its associated adverse reactions caused by ROT inhibition of mitochondrial complex I by activating cellular self-rescue mechanisms such as glycolytic pathways, TCA cycle and oxidative phosphorylation activities, and leads to new energy metabolism disorders and other NAD + The dependent biochemical process is disturbed, eventually leading to mitochondrial dysfunction, reduced energy production and ROS accumulation, leading to high oxidative stress. Similarly, astrocyte intracellular NAD + Level and NAD + The increase in NADH ratio favors its management of mitochondrial activity by enhancing glycolysis, TCA cycle and oxidative phosphorylationComplex I is not functional enough, so astrocytes with insufficient complex enzyme I function can maintain intracellular ATP levels and mitochondrial function homeostasis, thereby avoiding ROS accumulation and high oxidative stress and the deleterious reactions that result therefrom.
Accordingly, the effect of 100nM ROT exposure on the oxidative stress state of endothelial cells and astrocytes was further examined. The examination criteria included reduced coenzyme II (NADPH) and Glutathione (GSH) and their oxidized forms of NADP + And GSSG, and the level change of the ratio of the reduced form to the oxidized form, and the level change of the oxidative stress index Reactive Oxygen Species (ROS) and the index Mitochondrial Membrane Potential (MMP) of mitochondrial damage. NADPH and GSH are the two most important antioxidant substances in the body, and are critical to reduce intracellular ROS and prevent oxidative stress damage. When GSH scavenges ROS and is oxidized to GSSG, NADPH is required as an electron donor to reduce to GSH, thus GSH/GSSG and NADPH/NADP + The ratio of (2) and ROS levels can directly reflect cellular redox status and oxidative stress.
The results of the study are shown in table 53, with BMEC exposure to 100nm ROT for 36 hours significantly reduced intracellular GSH levels to 59% with nearly 6-fold increases in GSSG levels and 91% decreases in GSH/GSSG ratios, and a trend toward an increase in total glutathione (gsh+gssg), indicating that GSH levels decrease in association with synthesis but rather are impaired due to GSSG reduction to GSH. Consistently, NADPH/NADP + The equilibrium of (C) is also disrupted, whether NADPH and (NADPH+NADP) + ) Or NADPH/NADP + The ratio is significantly reduced, but NADP + The above results show that the level is not lowered but rather tends to rise, and the two points: first, NADPH production is reduced, and second, NADP + Cannot be timely reduced to NADPH, thus resulting in NADPH/NAPD + And the GSH/GSSG ratio is severely reduced. As a direct consequence of the severe reduction of these two ratios, ROS levels are significantly increased, while MMPs are significantly reduced. It can be seen that NADPH is produced and derived from NADP + The reduced regeneration results in reduced endothelial cell NADPH levels, which in turn results in reduced GSSG accumulation and GSH levels, elevated ROS levels, and mitochondrial damage. Consistent with the results of energy metabolism, ROT treatment under the same conditions did not significantly affect astrogliosis The indexes are examined, and the GSH/GSSG ratio tends to be increased, which indicates that astrocytes have the capacity to resist the initiation of NADPH production and NADP production from mitochondrial complex enzyme I insufficiency + Reduced regeneration, thus maintaining NADPH and NADPH/NADP + The steady state of the ratio, and thus the steady state of GSH and GSH/GSSG ratio, is maintained, avoiding redox imbalance and oxidative stress damage.
Table 53 endothelial cell redox balance is less plastic than astrocytes in response to mitochondrial complex enzyme I insufficiency
Figure BDA0004007353830000552
Figure BDA0004007353830000561
ROT: rotenone; GSH: reduced glutathione; GSSG: oxidized glutathione; gsh+gssg: the total amount of reduced glutathione and oxidized glutathione; GSH/GSSG: ratio of reduced glutathione to oxidized glutathione; NADPH: reduced nicotinamide adenine dinucleotide phosphate; NADP (NADP) + : oxidized nicotinamide adenine dinucleotide phosphate; NADPH+NADP + : total nicotinamide adenine dinucleotide phosphate; NADPH/NADP + : ratio of reduced to oxidized nicotinamide adenine dinucleotide phosphate; ROS, reactive oxygen species; MMP: mitochondrial membrane potential; ** p<0.01, *** p<0.001(vs.CON);n=5。
summarizing and discussing the above findings, ROT preferentially reduces vascular endothelial cell ATP and NAD + Levels, but elevated NADH levels, resulting in NAD + Serious imbalance of NADH ratio, NADPH/NADP + And redox homeostasis of GSH/GSSG is severely disrupted and mitochondria damaged. Whereas astrocytes increase NAD by increasing NAD + Level and NAD + The ratio of NADH is effective in treating mitochondrial complex enzyme I deficiency, so that energy metabolism, NADPH/NAD under conditions of mitochondrial complex enzyme I deficiency can be maintainedP + And redox homeostasis and mitochondrial function homeostasis of GSH/GSSG. This deeply explains the IC of ROT effect on endothelial cells 50 IC for astrocytes with a value of about 100nM 50 Scientific theory with a value of about 2000 nM. Therefore, improving the metabolic plasticity of neurovascular unit member cells, particularly endothelial cells, to the deficiency or absence of mitochondrial complex enzyme I function, particularly maintaining ATP, NAD + NADPH and NADPH+NADP + And GSH level, is critical to the prevention and the development of PD, and is a new idea for searching and finding anti-PD drugs with permanent cure effect. On this basis, applicants believe that it is by increasing this capacity of endothelial cells to cope with PDSC1 and other optimal compositions that eliminate the vulnerability of endothelial cells to deficiency or absence of complex enzyme I and that allow them to face mitochondrial complex enzyme I deficiency like astrocytes without onset. Thus, we design example 12-2 to further validate this speculation.
Example 12-2 PDSC1 maintains mitochondrial Complex enzyme I hypofunction or endothelial cell-deleted ATP and NAD + Level, redox homeostasis, mitochondrial function and prevention of oxidative stress damage.
Applicants have observed whether PDSC1 can increase the plasticity of endothelial cells to inhibit the energy metabolism and redox balance of mitochondrial complex enzyme I in response to Rotenone (ROT).
TABLE 54 plasticity of PDSC1 to enhance endothelial cell ability metabolism and redox balance in response to mitochondrial complex enzyme I dysfunction
Figure BDA0004007353830000562
ROT: rotenone; PDSC1: a panaxadiol saponin composition; ATP: adenosine triphosphate; ADP: adenosine diphosphate; ATP/ADP: the ratio of adenosine triphosphate to adenosine diphosphate; NADH: reduced nicotinamide adenine dinucleotide; NAD (NAD) + : oxidized nicotinamide adenine dinucleotide; NADH/NAD + : reduced nicotinamide adenine dinucleotide and oxidized nicotinamide adenine dinucleotideRatio of the dinucleotides; NADH+NAD + : the total amount of reduced nicotinamide adenine dinucleotide and oxidized nicotinamide adenine dinucleotide; GSH: reduced glutathione; GSSG: oxidized glutathione; GSH/GSSG: ratio of reduced glutathione to oxidized glutathione; gsh+gssg: the total amount of reduced glutathione and oxidized glutathione; NADPH: reduced nicotinamide adenine dinucleotide phosphate; NADP (NADP) + : oxidized nicotinamide adenine dinucleotide phosphate; NADPH/NADP + : reduced nicotinamide adenine dinucleotide phosphate; NADPH+NADP + : total nicotinamide adenine dinucleotide phosphate; ROS, reactive oxygen species; MMP: mitochondrial membrane potential; ** p<0.01, *** p<0.001(vs.ROT);n=5。
as shown in example 54, the results of the study demonstrate that PDSC1 can increase the metabolic plasticity of endothelial cells against the deficiency or absence of mitochondrial complex I, and thus can prevent mitochondrial dysfunction and subsequent cell damage caused by the deficiency or absence of complex I. Specifically, compared with a 100nM ROT injury group, the simultaneous administration of 25 mu M of PDSC1 can raise ATP level and ATP/ADP proportion, which suggests that the PDSC1 endows mitochondria complex enzyme I with low function or lacks endothelial cells to maintain oxidative phosphorylation of mitochondrial respiratory chain, thereby avoiding various malignant consequences caused by insufficient ATP. And the ATP level and ATP/ADP ratio were also slightly higher than those in astrocytes exposed to 100nM ROT (table 52). In particular, PDSC1 almost completely counteracts ROT-induced elevation of NADH levels and maintains NAD + Level and NAD + The NADH ratio is slightly higher than normal, so that endothelial cells with insufficient or absent function of the complex enzyme I have the ability to maintain energy metabolism homeostasis and ATP levels, thus avoiding ATP deficiency, NAD + Deficiency and NAD + Decreased NADH ratios lead to metabolic disorders and other pathogenic events.
Consistently, PDSC1 can significantly combat ROT-induced reduced levels of GSH and NADPH, GSSG and NADP + Elevated levels, elevated intracellular ROS levels, and reduced mitochondrial membrane potential. Furthermore, PDSC1 also significantly antagonizes the total amount of ROT-induced coenzyme II(NADPH+NADP + ) The reduction and significant increase in total glutathione (gsh+gssg) suggests that PDSC1 may increase the ability of complex enzyme I to function in deficient endothelial cells to synthesize both antioxidant substances. It can be seen that PDSC1 can promote not only NADPH and NADP + And GSH and GSSG are mutually converted to improve the capacity of endothelial cells with insufficient mitochondrial complex enzyme I function to remove ROS, and can strengthen the capacity of endothelial cells to disturb cell redox balance in response to the low mitochondrial complex enzyme I function by improving GSH synthesis. Furthermore, PDSC1 alone did not significantly affect the levels of the various indicators tested, indicating that PDSC1 does not interfere with normal energy metabolism and redox balance and thus PDSC1 is highly safe.
Taken together with the above results, it was demonstrated that PDSC1 can maintain NAD + Level and NAD + The steady state of NADH ratio and the redox balance mediated by NAPDH and GSH enhance the endothelial cells' ability to cope with mitochondrial complex I deficiency/depletion, thus allowing the endothelial cells to maintain near normal energy metabolism and ATP levels, redox balance and cell function status like astrocytes under conditions of complex I deficiency/depletion, thus avoiding oxidative stress injury and apoptosis of the ROT-induced endothelial cells and susceptibility of the diseased endothelial cells to ROT by other member cells of the neurovascular unit. Furthermore, PDSC1 does not interfere with normal energy metabolism and redox balance, indicating that PDSC1 is highly safe and further indicating that PDSC1 can selectively activate self-rescue ability in the face of endothelial cells with insufficient mitochondrial complex I function. Thus, the effect of PDSC1 is a specific expression of ginseng adaptation, and it is explained that PDSC1 and other excellent ginsenoside compositions can exert ginseng adaptation efficiently, and thus PDSC1 and other excellent ginsenoside compositions have a wide range of medical uses in increasing nonspecific resistance of the body against various endogenous and exogenous harmful factors such as physical (freezing, high temperature, excessive exercise, high pressure or low pressure), chemical (various toxic agents, narcotics, etc.), biological (xenogenic serum, bacteria, transplants, etc.).
Example 12-3. Maintenance of intracellular ATP levels by the preferred compositions is an important mechanism for combating the oxidative stress and inflammatory response induced by ROT.
Adenosine Triphosphate (ATP) deficiency, oxidative stress and inflammatory response are the resulting pathological events of mitochondrial dysfunction and are prevalent in the brain of neurodegenerative diseases, so they have been widely accepted as drug targets for protecting nerves and controlling related brain diseases. However, the underlying links of these three pathological events are not yet clear. Thus, revealing the intrinsic links of ATP deficiency, oxidative stress, and inflammatory responses may further demonstrate the mechanism by which PDSC1 increases endothelial cell response to mitochondrial complex enzyme I deficiency, and further reveal the common mechanism by which PDSC1 and other optimal compositions control neurological diseases including PD and AD and other mitochondrial dysfunction diseases. Accordingly, the applicant observes the effect of each on ROT-induced apoptosis by adding exogenous ATP, GSH and NADPH, respectively, and 100nM ROT simultaneously with endothelial cells, and the results of the study make it clear that the three pathological events described above have a relative pathological role in this model. As shown in table 55, exogenous ATP, GSH, and NADPH all significantly reduced ROT-induced endothelial apoptosis. It can be seen that both ATP deficiency and oxidative stress associated with GSH and NADPH deficiency are involved in ROT-induced apoptosis.
Table 55 addition of exogenous ATP, GSH and NADPH all significantly counteracts ROT-induced mitochondrial endothelial apoptosis
Figure BDA0004007353830000571
Figure BDA0004007353830000581
ATP, adenosine triphosphate; ROT: rotenone; GSH: reduced glutathione; NADPH: reduced nicotinamide adenine dinucleotide phosphate; *** p<0.001 (vs. ROT group); n=5.
Next, it was observed that exogenous ATP can combat ROT-induced oxidative stress, inflammatory response, and cell damage? The experimental results are shown in Table 56, with the addition of exogenous ATPGSH, GSSG, NAD which can significantly counteract ROT induction + 、NADH、NADPH、NADP + And their respective reduced and oxidized forms of the relative content of changes and elevated ROS levels, and against ROT-induced endothelial cells to release pro-inflammatory factors. In particular, exogenous ATP can significantly inhibit ROT-induced increase in NADH levels and NAD + reduced/NADH level and maintenance of NAD + The NADH ratio was near normal (0.94), which was similar to that of the ROT control group + A strong contrast is produced by decreasing the NADH ratio to 0.21. This suggests that in endothelial cells with insufficient mitochondrial complex enzyme I function, ATP deficiency results in NADH to NAD + A reduction in conversion indicative of inhibition/dullness of glycolytic and respiratory chain electron transfer activities, which in turn exacerbates ATP level reduction, while NADH is directed to NAD + The reduction in conversion also means NAD + The dependency has a low function and even suffers from a loss. As can be seen, ATP levels are compared with NAD levels and NAD + There is a close relationship between NADH ratio, insufficient availability of ATP and NAD + the/NADH ratio imbalance can be causal to each other and, together, disrupt NADPH and GSH mediated redox balance, oxidative stress and pro-inflammatory states. Because multiple metabolic enzymes in the tricarboxylic acid cycle and respiratory chain are extremely sensitive to ROS, GSH deficiency can in turn exacerbate ATP deficiency and its malignant consequences. In addition, GSH is a functionally important molecule, including antioxidants, regulators of DNA synthesis and repair, protectants for thiol groups in proteins, stabilizers for cell membranes, antidotes for exogenous substances, etc., and GSH deficiency will fully impair cell function and promote cell structure degeneration and cell death. This further illustrates that "maintaining ATP levels and moderately increasing NAD + Level and NAD + the/NADH ratio "is a key mechanism by which astrocytes have a strong ability to cope with ROT cytotoxicity than endothelial cells, and PDSC1 is a mechanism by which to improve the ability of endothelial cells to cope with insufficient complex enzyme I functions. Furthermore, PDSC1 can increase the ability of mitochondrial complex I-deficient endothelial cells to scavenge ROS by promoting NADP and GSSG back to reduced form and increasing GSH synthesis.
Table 56 exogenous ATP protects endothelial cell redox balance against the impact of complex I deficiency and inhibits inflammatory response
Figure BDA0004007353830000582
ATP, adenosine triphosphate; ROT: rotenone; GSH: reduced glutathione; GSSG, oxidized glutathione; GSH/GSSG: ratio of reduced glutathione to oxidized glutathione; gsh+gssg: the total amount of reduced glutathione and oxidized glutathione; NADPH, reduced nicotinamide adenine dinucleotide phosphate; NADP (NADP) + Oxidized nicotinamide adenine dinucleotide phosphate; NADPH/NADP + Reduced nicotinamide adenine dinucleotide phosphate; NADPH+NADP + : total nicotinamide adenine dinucleotide phosphate; NADH: reduced nicotinamide adenine dinucleotide; NAD (NAD) + : oxidized nicotinamide adenine dinucleotide; NADH/NAD + : ratio of reduced nicotinamide adenine dinucleotide to oxidized nicotinamide adenine dinucleotide; NADH+NAD + : the total amount of reduced nicotinamide adenine dinucleotide and oxidized nicotinamide adenine dinucleotide; ROS: active oxygen; TNF- α: tumor necrosis factor alpha; IL-1. Beta: interleukin 1-beta; IL-6: interleukin 6; *** p<0.001(vs.ROT);n=5。
summary and discussion:
(1) PDSC1 activates cellular self-rescue mechanisms and protects them from the biochemical parenchyma of ROT toxicity.
Comprehensive example 12 results demonstrate that intracellular ATP and NAD during ROT-induced endothelial cytotoxicity + Deficiency and NAD + Serious imbalance of the NADH ratio leads together to depletion of NADPH and GSH and destruction of the cell's antioxidant function. Conversely, disruption of antioxidant function exacerbates ATP and NAD deficiency to the extent of depletion by failing to protect the TCA cycle and the respiratory chain from multiple metabolic enzymes extremely sensitive to ROS, ultimately leading to ROS accumulation, oxidative stress damage, inflammatory reactions, mitochondrial damage, and cell death. In strong contrast to endothelial cells, the mitochondrial complex enzyme is induced in the face of ROTWhen the I function is insufficient, the astrocyte can moderately improve the NAD + Level and NAD + NADH ratio, which provides a prerequisite for the cells to successfully combat the effects of complex enzyme I insufficiency on ATP production by increasing glycolysis and TCA cycle activity, thus maintaining intracellular ATP levels at adequate levels and NADPH and GSH mediated redox homeostasis, thereby avoiding ROS accumulation, mitochondrial damage and the appearance of apoptosis. This reasonably explains the IC of the effect of ROT on astrocytes 50 The values are IC's acting on endothelial cells 50 The value was approximately 20 times. It can be seen that intracellular ATP and NAD are maintained + At a sufficient level and with a suitable NAD + The NADH ratio is critical for preventing and treating mitochondrial complex enzyme I insufficiency and other PD risk factors and even various risk factors of mitochondrial dysfunction or injury from causing diseases. Indeed, PDSC1 moderately increases NAD for endothelial cells exposed to ROT toxicity + And NAD + NADH ratio and maintains ATP levels at sufficient levels and promotes regeneration of NADPH and GSH from its oxidized form and increases GSH synthesis, thus not only maintaining but also enhancing NAPDH and GSH mediated cellular antioxidant function, so ROT-induced oxidative stress injury, inflammatory response and apoptosis of endothelial cells can be avoided. Taken together, the results of the study revealed that endothelial cells are vulnerable to ROT inhibiting mitochondrial complex I function and PDSC1 activates cellular self-rescue mechanisms to protect against the biochemical essence of ROT toxicity.
In particular, mitochondrial dysfunction (with ATP deficiency), oxidative stress injury, and neuroinflammation are common pathological events of various neurodegenerative diseases including PD, AD, HD, and ALS. However, none of the clinical trials of antioxidants and anti-inflammatory agents or actives to treat PD and other neurodegenerative diseases have achieved satisfactory results. We have found that ATP and NAD are absent in the pathological course of neurodegenerative diseases + And NAD + Deregulation of the NADH ratio is an upstream pathological event that PDSC1 can avoid or correct, and thus is effective in the prevention of PD and other neurodegenerative diseases. The efficacy of PDSC1 and other potent compositions in ROT-induced PD rat models is well documented for their effects on PD genesis and other conditionsThe effectiveness and high safety of development. In the following examples, we will conduct further validation studies.
(2) The research results also support the wide medical application of the PDSC1 and other excellent ginsenoside compositions in preventing and treating neurodegenerative diseases, mitochondrial diseases, aging-related diseases and delaying aging.
Mitochondrial dysfunction is accompanied by energy (ATP) deficiency, oxidative stress damage, and neuroinflammation, common pathological events of various neurodegenerative diseases including Parkinson's Disease (PD), alzheimer's Disease (AD), huntington's Disease (HD), and Amyotrophic Lateral Sclerosis (ALS), which are also important causes of accelerated aging. Glutathione (GSH) is the most abundant non-protein thiol, playing a vital role in the antioxidant defense system and maintenance of neuronal redox homeostasis. Thus, GSH has a number of important functions, including antioxidants, regulators of DNA synthesis and repair, protectants for thiol groups in proteins, stabilizers for cell membranes, and antidotes for exogenous substances. The levels of GSH in the brain also play a critical role in preventing ischemia/stroke and maintaining blood brain barrier integrity. It will be appreciated that the lack of GSH in the brain is involved in the occurrence and development of neurological diseases such as PD, AD and HD, and is also associated with neuronal loss during aging. Oxidative stress and GSH deficiency are also closely related to neurovascular disorders (including stroke and diabetic retinopathy) and psychotic disorders (including substance abuse, autism, obsessive-compulsive disorder, schizophrenia, depression, bipolar disorders). Therefore, prevention and treatment of neurodegenerative diseases and other GSH deficiency-related diseases by addition of GSH precursor amino acids is widely appreciated. NAD (NAD) + Is a further multifunctional molecule, NAD + Lack is another common feature of neurodegenerative diseases and aging. NAD (NAD) + As redox coenzymes are the center for energy metabolism, glycolysis, TCA cycle and mitochondrial oxidative phosphorylation all rely on sufficient NAD + And a suitable NAD + NADH ratio. Furthermore, NAD is required for many key enzymes regulating various cellular functions such as protein stabilization from the gene expression + As co-substrates for their catalytic activity, these enzymes include NAD + Dependency ofProtein deacetylase sirtuin family and DNA repair enzyme PARP family, etc., thus NAD + Many other critical cellular functions are also involved, including DNA repair, chromatin remodeling, mitochondrial homeostasis, and cellular aging. NAD (NAD) + Deletions lead to mitochondrial dysfunction, reduced energy production and ROS accumulation, ultimately leading to high oxidative stress and cell senescence and death. NAD of the human brain and other organs with age + The level drops. There is increasing evidence that NAD + Steady state imbalance and NAD + Imbalance in the NADH ratio can lead to a variety of disease states including neurodegenerative disorders (PD, AD, HD and ALS) and other age-related diseases, NAD + The lack also accelerates the aging process. Mitochondrially producing ATP to support neural activity, including neurotransmission and Ca 2+ Homeostasis, and is a signal source regulating nuclear mitochondrial communication and even neuronal survival and death arbitration. Thus, protecting mitochondrial function, antioxidant, anti-inflammatory and enhancing GSH and NAD by supplementing precursor substances + Is considered as a reasonable approach for treating neurodegenerative diseases and delaying aging, however, no satisfactory effect has been obtained in the related clinical trials so far. Our findings indicate that ATP and NAD are available during complex enzyme I inhibitor Rotenone (ROT) -induced oxidative stress, inflammatory response and subsequent cell death + Deficiency and concomitant NAD + The decreasing imbalance of NADH ratio is an upstream event leading to NADPH and GSH deficiency, and ATP and NAD + The deficiencies may be causal to each other. Further, an important direct consequence of NADPH and GSH deficiency is ROS accumulation, which in turn causes inflammatory reactions and extensive oxidative damage, where the damaged TCA cycle and metabolic enzymes in the respiratory chain can further exacerbate ATP and NAD + Deficiency and mitochondrial dysfunction. Astrocytes moderately increase NAD + And NAD/NADH ratio and maintaining ATP levels at a sufficient level for use, which is resistant to IC of the Complex enzyme I inhibitor ROT 50 The value is approximately 20 times that of endothelial cells. It can be seen that ATP and NAD are maintained or recovered + Level and NAD + The NADH ratio is a more effective agent in physiological range than an antioxidant and anti-inflammatory agent or a functional molecule targeting mitochondriaNew strategies for systemic degenerative diseases and for delaying aging. PDSC1 can implement this new strategy to maintain and protect energy metabolic homeostasis, redox homeostasis, mitochondrial function, and NAD from the source + GSH and NADPH mediated various physiological functions. Furthermore, PDSC1 may also promote the regeneration of NADPH and GSH from its oxidized form and increase GSH synthesis to enhance the ability of mitochondrial complex I-deficient endothelial cells to scavenge ROS. The cytotoxin which protects endothelial cells, astrocytes and nerve cells against ROT is a functional feature of the PDSC1 described above, and it is expected that the combination of PDSC1 with precursor amino acids of NAD and GSH (including serine, glycine and glutamine) can obtain better resistance against NAD causing diseases + And GSH deficiency effects. In conclusion, the PDSC1 and other effective ginsenoside compositions have wide application prospects in preventing and treating neurodegenerative diseases, neurovascular diseases (including stroke and diabetic retinopathy) and mental disorder diseases and delaying aging.
The research result also supports the medical application of the PDSC1 and other excellent ginsenoside compositions in treating and preventing mitochondrial diseases. Mitochondrial diseases are a group of heterogeneous diseases caused by defects of mitochondrial metabolic enzymes caused by genetic defects, such as ATP synthesis disorder and energy source deficiency, including mitochondrial myopathy and mitochondrial encephalomyopathy, and no specific drug treatment is currently carried out on the diseases, and Adenosine Triphosphate (ATP), coenzyme Q10, a large amount of B vitamins and the like are mainly fed. Therefore, our findings predict the medical use of PDSC1 and other superior ginsenoside compositions alone or in combination with ATP, coenzyme Q10 and B vitamins to treat mitochondrial disease.
(3) The research results support the medical application of the PDSC1 and other excellent ginsenoside compositions in treating the brown skin disease.
Piano disease, also known as niacin deficiency, also known as niacin or niacin deficiency, is clinically manifested by symptoms of the skin and nervous system (most commonly seen in neurasthenia, and also manifested as mental symptoms and dementia, and also peripheral neurological symptoms such as multiple peripheral neuritis). Occurrence of brown leather disease and NAD + Reduced uptake and absorption of synthetic precursor substances such as niacin and tryptophan and associated metabolic disorders. As can be seen, applicants' studies have found that increased understanding of the pathogenesis of brown skin disease more predictive of PDSC1 and other superior ginsenoside compositions alone or in combination with NAD + Synthetic precursor substances such as tryptophan, nicotinic Acid (NA), nicotinamide (NAM), nicotinamide Riboside (NR) and Nicotinamide Mononucleotide (NMN) and NAD + The medical application of synthesizing the required vitamins for preventing and treating the pellagra is provided.
(4) The research result reveals the effect of the PDSC1 on improving the cell adaptability steady-state function, and further supports the medical application of the PDSC1 and other excellent ginsenoside compositions for delaying aging and the medical application for improving sub-health state and preventing and treating stress diseases.
Adaptive homeostasis (or adaptive homeostasis) is the "transient expansion or contraction of the homeostatic range of any given physiological parameter in response to exposure to sub-toxic, non-destructive, signaling molecules or events, or removal or cessation of such molecules or events". Adaptive homeostasis enables continuous short-term adjustments of biological systems in constantly changing internal and external environments to achieve optimal function. Adaptive initiation of appropriate signals enables organisms to successfully cope with larger, often toxic stressors (such as hypoxia, oxidative stress, immune response and psychological stress of various origins, etc.). However, adaptive homeostasis, i.e. the ability to cope with momentary changes in internal and external environmental pressure, diminishes with age, especially in the last third of life. Studies on cultured mammalian cells, worms, flies, rodents, apes and even humans have shown that a decrease in adaptive homeostasis is a hallmark of aging and predisposes the elderly to a variety of diseases including neurodegenerative, cardiovascular and diabetic diseases, and the like. Reduced adaptive homeostasis also reduces resistance to a variety of stressors, including somatic, social, psychological and mental cultural stressors, and thus the elderly are vulnerable to a variety of internal and external environmental stimuli. It is also specifically pointed out that for people of other ages sub-health conditions (e.g. sleep disorders, reduced ability to work with negative emotions, brain and physical forces) and stress related diseases (negative emotions inducing and aggravating essential hypertension; stress related mental disorders including acute cardiac responses, delayed cardiac responses and adaptation disorders; inflammation related diseases such as heart diseases and autoimmune diseases; immunosuppression related diseases such as responses to viral infections and vaccination) are also caused when the stress causes a stress exceeding the stress that can be sustained by the adaptive steady-state function of the body, even directly caused (also called stress disorders such as stress ulcers; post-traumatic stress disorders, delayed appearance and prolonged lasting mental disorders due to threatening, catastrophic psychological wounds, etc.), especially individuals with small adaptive homeostatic elastic space (poor plasticity) due to genetic and physical factors are vulnerable to various stress. As can be seen, successfully maintaining adaptive homeostasis below the upper physiological limit not only improves the quality of life and prolongs the life of the elderly, but also relieves the young and young sub-health states caused by various stresses and reduces stress-related diseases.
Based on the NAD + Is believed to be sufficient intracellular ATP and NAD + Level and suitable NAD + NADH ratio is critical to maintaining cell adaptive homeostasis, ATP and NAD + Depletion then impairs the cell-adaptive stress response and thus leads to stress injury. Three major contributors to neuronal adaptive cellular stress responses are ATP consumption, ROS production, and Ca 2+ A signal. With age, these stress responses make neurons more susceptible to various forms of endogenous or exogenous stress. We have further found that for the same stressor, the mitochondrial coenzyme I inhibitor ROT exposure (leading to mitochondrial complex enzyme hypofunction/loss), vascular endothelial cells are stressed with NAD + Depletion, ATP depletion, depletion of antioxidant substances GSH and NADPH and their oxidized form accumulation, ROS production, mitochondrial damage and apoptosis; whereas astrocyte stress is a moderate increase in NAD + Level and NAD + NADH ratio, maintenance of ATP at sufficient levels, redox homeostasis of GSH and NADPH, absence of ROS elevation, mitochondrial damage and apoptosisAnd (3) death. Furthermore, ATP depletion and NAD depletion + Consumption can be causal, ATP consumption can lead to NADPH and GSH consumption and inflammatory reactions. Consistently, the IC of ROT effect on astrocytes 50 The values are IC's acting on endothelial cells 50 Values of approximately 20 times, but diseased endothelial cells greatly reduce the vulnerability of astrocytes to ROT by releasing inflammatory factors and severely impair the function of astrocytes to maintain protein homeostasis by phagocytic activity while incurring the neurotoxicity of the astrocyte A1 phenotype. Taken together, intracellular ATP and NAD are maintained + At sufficient levels, especially moderately elevated NAD + Level and NAD + The NADH ratio is critical to maintaining cell adaptive homeostasis. Chronic oxidative stress and defective protein homeostasis such as aβ and deposition of α -synuclein are two major causes of age-related diseases. Aging is characterized by increased oxidative stress and reduced adaptive steady state response, thus resulting in accumulation of oxidative damaged proteins, DNA and lipids in the cell. As can be seen, our findings reveal the biochemical mechanisms of chronic oxidative stress and defective protein homeostasis during aging, and the findings that endothelial cells are vulnerable and vulnerable to adaptive homeostasis with insufficient mitochondrial complex enzyme I function also reasonably explain the high incidence of brain microvascular damage commonly found in neurodegenerative diseases and cerebrovascular diseases in the elderly population. More importantly, our findings reveal that by targeting intracellular ATP and NAD + A novel approach for maintaining or even improving the function of adaptive homeostasis of cells, which has wide application prospects in delaying aging, and preventing and treating stress diseases and neurodegenerative diseases.
The PDSC1 and other effective ginsenoside composition can exert the medical purposes of delaying aging, improving the survival quality of the old and preventing and treating stress diseases and neurodegenerative diseases through the novel way. PDSC1 depletes endothelial cell ATP against ROT exposure, disrupts the redox balance of NADPH and GSH, activates inflammatory pathways, and increases NAD to some extent + Level and NAD + NADH ratio. Accordingly, PDSC1 protects endothelial cells, astrocytes and neurons from toxic exposure to ROTThe method comprises the steps of carrying out a first treatment on the surface of the In the ROT-induced PD rat model, PDSC1 almost completely blocks the occurrence of ROT-induced PD clinical motor symptoms, neurotoxic A1 phenotype, α -synuclein deposition, and neuroinflammation, and protects neurovascular units in an almost normal state. It is also specifically noted herein that PDSC1 does not interfere with the level of the biochemical indicators described above in the normal state of the cell. It can be seen that PDSC1 does not disrupt adaptive homeostasis of cells, but rather that the mechanism of self-rescue to selectively activate endothelial cells facing deficient mitochondrial complex I function involves modest increases in NAD + Level and NAD + the/NADH ratio (thus maintaining ATP at a sufficient level), promotes regeneration of NADPH and GSH from its oxidized form and increases GSH synthesis (thus increasing the ability to scavenge ROS), thus increasing the ability of endothelial cells to cope with otherwise deleterious stressors (mitochondrial complex enzyme insufficiency/deficiency), and further increasing the damage threshold of ROT to astrocytes and neurons. Ginseng has been widely accepted as an adaptation-like effect (increasing the body's nonspecific resistance, i.e., ginseng can enhance body's resistance to adverse effects of various harmful factors such as physical (freezing, high temperature, excessive exercise, high pressure or low pressure), chemical (various toxic agents, narcotics, etc.), biological (xenogeneic serum, bacteria, transplanted tumors, etc.), and this effect is considered as a substance of ginseng's efficacy of strengthening body resistance. The medicine can promote the transformation of the pathological changes of the organism into the normal physiological state to achieve the boundaries of 'yin-yang-secret and spirit-treatment'. Therefore, only when the body resistance is reduced or the body is overloaded, the medicine can exert obvious curative effect, but under normal conditions, the medicine effect is often not obvious, namely the medicine is ' deficiency, supplement and the medicine is ' deficiency '. Obviously, the effect of the PDSC1 on selectively activating endothelial cell self-rescue mechanism to cope with toxic ROT exposure fully reflects the effect of ginseng on adapting to the original shape/strengthening body resistance and consolidating constitution. Dopamine 2 receptor haloperidol (HAL, used for acute and chronic schizophrenia, mania and tourette syndrome) is also a stressor for the body when It is the side effect that we have found that PDSC1 and other preferred ginsenoside compositions can slow down the motor symptoms of PD by a mechanism independent of dopamine, which not only reveals that PDSC1 and other preferred compositions differ from L-DOPA and improve late PD motor symptoms insensitive to L-DOPA, but also verifies from another point of view that PDSC1 can exert ginseng adaptation as such.
(5) The research results support the medical application of the PDSC1 and other optimal compositions for preventing and treating peripheral neuropathy.
Peripheral Neuropathy (PN), a disease caused by structural and functional dysfunction of peripheral motor nerves, sensory nerves and autonomic nerves, is a common disease in clinical practice, and has constituted a major public health problem. The peripheral nerves innervate the five sense organs, the movements and sensations of the trunk of the extremities, and the sensory abnormalities (needle sticks, burns, pain, etc.) caused by peripheral nerve damage from external or internal causes, dyskinesias, and impaired or lost function (or twitches, or muscle weakness, which may be accompanied by muscle atrophy), are collectively referred to as peripheral neuropathy. The peripheral neuropathy is commonly caused by trigeminal neuralgia, facial neuritis, facial spasm, glossopharyngeal neuralgia, occipital neuralgia, sciatica, intercostal neuralgia, multiple peripheral neuropathy, guillain-Barre syndrome and other diseases, wherein the trigeminal neuralgia, facial neuritis and facial spasm are most common in clinic. Common causes of peripheral neuropathy include trauma, diabetes, neurotoxic chemotherapy, viral infections (e.g., post-herpetic neuralgia and HIV-induced peripheral neuropathy), alcoholism, nutritional deficiencies, heavy metal toxicity, neuronal cell autoradiography (e.g., demyelination, axonal lesions), and the like. Although the causes of initiation are diverse, more and more studies have demonstrated that mitochondrial dysfunction is the primary pathological mechanism of peripheral neuropathy. Specifically, mitochondrial dysfunction-mediated ATP deficiency, oxidative stress, and inflammatory reactions and apoptosis are all involved in the initiation and progression of peripheral neuropathy. Since axons are quite long, the peripheral nerves have special energy requirements, so proper mitochondrial function and distribution along the nerves is critical. Thus, pharmacological modulation of mitochondria, either directly or indirectly, is believed to be expected to produce therapeutic relief from a variety of primary and secondary mitochondrial diseases. It is also noted herein that inflammatory factor mediated neuroinflammation plays an important role in polyneuropathy, postherpetic neuralgia, and fibromyalgia syndrome. In brain oxidative stress injury is accompanied by neuroinflammation, we found that energy (ATP) deficiency caused by mitochondrial dysfunction induces oxidative stress injury and inflammatory response. Thus, it is reasonable to think that neuroinflammation is also involved in other peripheral neuropathy. Based on the important role of mitochondrial dysfunction-related metabolic disorders and neuroinflammation in peripheral neuropathy, the removal of metabolic disorder toxic products and inflammatory factors from the blood by therapeutic apheresis/therapeutic plasmapheresis is considered a new effective treatment. The current anticonvulsants gabapentin and pregabalin are widely used clinically for the treatment of peripheral neuropathy caused by various causes due to their pharmacological actions of blocking presynaptic terminal voltage-gated sodium and calcium channels and thus down-regulating the release of excitatory neurotransmitter Glu. In addition, vitamin B1, adenosylcobalamin, mecobalamin, and the like, which are considered to be capable of performing trophic nerve treatment, are also commonly used for treating peripheral neuropathy caused by various causes.
Compared with the prior medicines and technologies, the PDSC1 has outstanding pharmacological effects, and is characterized by protecting mitochondrial function, maintaining the steady state of energy metabolism and redox balance and preventing neuroinflammation. It can be seen that PDSC1 can prevent the onset and inhibit the progression of disease against the central mechanism of peripheral neuropathy and its multiple key nodes downstream. It is also noted herein that schwann cells (schwann cells) are glial cells in the peripheral nervous system that are distributed along the processes of neurons, also coat axons/nerve fibers to form myelin and promote nerve signaling, which also secrete neurotrophic factors, promote survival of damaged neurons and regeneration of their axons. Microvascular lesions and damaged schwann cells are considered to be important causes of peripheral neuropathy. There is increasing evidence that there is a complex interaction between schwann cells, axons and capillaries, and that this interaction is disrupted leading to diabetic neuropathy. Microvascular damage may disrupt its function by inflammatory cascades and by disrupting the pathways of schwann cells to gain oxygen and glucose, whereas oxidative stress and mitochondrial disturbances in diseased schwann cells may lead to dysfunction of neurons. It can be seen that the relationship among vascular endothelial, schwann cells and nerve fibers at the periphery is quite similar to that of endothelial cells, astrocytes and neurons in the neurovascular units in the brain found by the applicant. Therefore, the same efficacy of PDSC1 and other effective compositions for peripheral vascular endothelial, schwann and nerve fibers and their interactions can be speculated from the role of PDSC1 in protecting each member cell of the neurovascular unit and in breaking the pathological link between each member cell.
In summary, PDSC1 and other advantageous compositions may provide a novel therapeutic approach consistent with peripheral neuropathy pathogenesis. Due to the different principles of action, PDSC1 and other optimal compositions may also be used in combination with existing therapeutic agents or neurotrophic therapies. Finally, in the subsequent researches, the related embodiments will be further provided for the medical application of the PDSC1 and other effective ginsenoside compositions in delaying aging, improving the life quality of the aged, preventing and treating stress diseases and neurodegenerative diseases, and relieving the toxic and side effects of clinical drug treatment.
Example 13. Optimal compositions protect against PD striatal neurochemical homeostasis/PDSC 1 correction of late PD rat striatal neurotransmitter disorders and against L-DOPA chronic treatment disturbing neurotransmitters.
Parkinson's Disease (PD) is a progressive chronic disease, and no drug has been known to delay its progression. Usually, when a patient is clinically diagnosed with PD, more than 70% of dopamine neurons in the substantia nigra pars compacta have been lost, and as dopamine neurons continue to be lost, motor symptoms and non-motor symptoms of PD are aggravated. The classical PD pathology is Dopamine (DA) deficiency and thus replacement therapy with L-DOPA, but its remarkable expiration period is only 3 to 5 years, and then not only gradually decreases in efficacy but also develops late motor symptoms including gait freezing and posture impairment (leading to frequent falls), and disabling side effects including catabolism, delusions hallucinations, dementia, etc. More and more studies have demonstrated that the normal activity of acanthose neurons (SPNs) of the striatum, in addition to Dopamine (DA), is regulated by a variety of neurotransmitters including glutamate (Glu) and gamma-aminobutyric acid (GABA) and is involved in the expression of PD symptoms and disease progression. The involvement of Glu hyperexcitability in the expression of motor symptoms of PD in the period of DA signal shutdown is also closely related to the onset of catabolism caused by L-DOPA therapy (non-physiologic opening of DA signals), and it is apparent that continuous hyperexcitability of Glu will lead to degeneration of the peripheral and interneurons of various afferent nerves in the striatum and promote disease progression. Striatal GABA dysfunction is also an important component of PD disease, and is closely related to PD motor dysfunction (especially dyskinesias and bradykinesia) and neuropsychiatric behavioral disorders (such as cognitive dysfunction, depression, apathy, anxiety, etc.). However, abnormal increases in striatal GABA levels during the period of DA signal shutdown are closely related to the occurrence of bradykinesia in PD patients. It can be seen that maintaining and reconstructing striatal Glu and GABA homeostasis is critical for slowing PD motor symptoms and disease progression. The studies of the previous examples found that both the effects of PDSC1 (example 10) including almost complete resistance to ROT-induced PD rat striatal PV positive interneuron loss (example 11) and astrocyte loss (function to protect it from reducing extracellular Glu levels) and the effects of PDSC1 to enhance adaptive homeostasis of cells (example 12) were predicted to both maintain or reconstruct Glu and GABA homeostasis of the PD striatum and to act against L-DOPA-disturbed striatal neurotransmitters by PDSC 1. Accordingly, applicants used 6-hydroxydopamine (6-OHDA) to induce a mid-late stage PD rat model, and studied the effects of PDSC1, L-DOPA, and a combination of both on the PD striatal neurochemical microenvironment in comparison to verify that PDSC1 has the effect of modulating PD striatal neurochemical homeostasis.
Research method
And (3) model preparation: the unilateral injury middle and late PD rat model was induced by injecting 6-OHDA (12. Mu.g 6-OHDA in 4. Mu.L of physiological saline containing 0.01% ascorbic acid) into the unilateral forebrain medial bundle using conventional methods. The injection site positioning coordinates were: relative to forebrain and dural surface (AP: -1.8mm; ML: +2.0mm; DV: -8.6 mm). Intraperitoneal injection of 2mg/kg amphetamine on day 15 of 6-OHDA injection induced rotational behavior to examine whether molding was successful, with no less than 5 rotations per minute being considered successful.
Grouping and administration: the experiments were carried out with a normal animal control group, a PD model group, a Medoba group (23 mg L-DOPA+5.75mg benserazide/kg, hereinafter referred to as L-DOPA for short, 7 times the dose per kg of human), a PDSC1 group (20 mg/kg) and a PDSC1+L-DOPA group. The administration is carried out once a day for 21 continuous days, the brain microdialysis test is carried out on the 22 th day (36 th day of modeling), and brain specimens can be obtained after microdialysis is finished.
Microdialysis of awake animals: the microdialysis catheter was implanted under anesthesia at the dorsal striatum part (AP: +0.6mm; ML:3.5mm; DV: -6.0 mm) on the lesion side, and dialysate samples were collected with a microdialysis probe (4 mm) at a set period of time, and were sampled at low temperature (-20 ℃) every 20 minutes at a dialysis rate of 0.5. Mu.L/min, including 5 samples per animal for baseline samples. After each sample collection, the dialysate was flash frozen and stored at-80 ℃ for later use.
Microdialysis method: numerous studies have demonstrated that the optimal time period for performing microdialysis experiments is 18-48 hours after the probe catheter is installed, since then tissue damage around the probe has been restored, and after three days dialysis is affected by gliosis around the dialysis catheter. Thus, all microdialysis experiments of this study will be completed within 18-24 hours of implantation of the catheter. While the catheter is being implanted, a probe (2 mM) is inserted, and artificial cerebrospinal fluid (NaCl 147mM, KCl 3.5mM, mgCl) is used 2 1.2 mM、CaCl 2 1.2 mM、NaH 2 PO 4 1.0 mM, pH 7.0-7.4) the smoothness of the dialysis system was maintained at 0.3. Mu.L/min (the flow rate was increased to 1.5. Mu.L/min when the dialysate was collected formally the next day), and the animals were kept in a freely movable device box overnight. Typically the catheter was set in the afternoon, and rats were then acclimatized overnight in a microdialysis cage for awake animals. During microdialysis, the pump speed was set at 1.5. Mu.L/min and equilibrated for 2 hours. Then, 15. Mu.L of the dialysate was collected every 20 minutes in a sample tube containing 1. Mu.L of 1.5M acetic acid at-20 ℃. After sample collection, the sample was collected by reverse dialysis with 3% bromophenol blueThe position of the probe was verified and the data were analyzed using only animals with the probe correctly located on the dorsal side of the striatum.
Glu and GABA neurotransmitter assays: glu and GABA levels were determined using a laboratory established liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) method. LC-MS/MS analysis was performed on a Shimadzu-30A system and AB-QD 4500 mass selective detector using Agilent 120EC-C 18 Porosill column (100X 2.1mm,2.7 μm) with a sample loading of 1. Mu.L. The mobile phase (solvent A: acetonitrile; solvent B: 0.1% formic acid in water) is transported at a flow rate of 350. Mu.L/min and eluted with a linear gradient (0.0 to 5.0min,2 to 30% A;5.0 to 5.1min,30 to 90% A;5.1 to 5.3min,90% A;5.3 to 5.6min,90 to 2% A;5.6 to 8.0min,2% A). The selected parameters for its determination are shown in table 57.
Table 57LC-MS/MS determination of parameters of neurotransmitters Glu and GABA
Figure BDA0004007353830000631
Substantia nigra compact part survival dopamine neuroanalysis: the deeply anesthetized rats were perfused with a conventional fixative solution through the heart to fix brain tissue, and the brain segments were collected by a conventional method with continuous coronal sections (40 μm) of the substantia nigra section (from bregma-4.52 mm to-6.04 mm) and semi-quantitatively required, respectively. Briefly, brain regions containing black matter were collected from 10, 20 and 35 sections from each animal for quantitative analysis, with the average of 3 sections as data for one animal. The density of TH positive neurons in the substantia nigra pars compacta was observed by immunohistochemical method to confirm the survival of dopamine nerves.
Study results 1: changes in extracellular Glu and GABA of the striatum of PD animals and chronic therapeutic effects of L-DOPA.
In order to find the intrinsic link of a possible PD striatal neurotransmitter disorder with a dopamine receptor dysfunction, it is necessary here to understand: dopamine receptor subtype 1 (D1R) located on the lateral side of spiny neurons (SPNs) promotes GABA release from the lateral terminals, while D2R inhibits GABA release, and D2R also inhibits Glu release from the cortical afferent Glu-capable nerve terminals.
As shown in table 58, the extracellular Glu and GABA levels were increased by 6.01 and 2.97 fold, respectively, in the 6-OHDA model group rats compared to the normal group animals, indicating a significant increase in Glu/GABA ratio, indicating an increase in Glu excitability in the PD striatum. The L-DOPA chronic treatment animals, at the dosing interval, 24 hours after the last dose and before the current dose (i.e., dopamine signal "off" period), had significantly higher Glu and GABA levels than the model animals, and had slightly greater lift levels of Glu than the rise of GABA, so that the Glu/GABA ratio was further deviated from normal levels, indicating that chronic L-DOPA dosing further worsened the excitability and inhibitory imbalance of the striatum during the "off" period.
Elevated striatal Glu levels during the DA "on" phase are known to be closely related to L-DOPA-induced catabolism. Thus, the function of DA was turned on with L-DOPA, and the changes in extracellular Glu and GABA levels of the striatum were compared to those of model animals and chronically administered L-DOPA. Experimental results indicate that the two groups of animals show great differences in response to acute stimulation by L-DOPA. In the model group, animals developed a decrease in Glu levels at 20 minutes (70.66% of basal), followed by a gradual increase, peaking at 120 minutes (table 59), the Glu area under the curve being significantly higher than the control group animals (table 60); and the GABA level continuously decreases until the GABA level rises within 120min, and the curve area of the GABA is reduced remarkably more than that of animals in a control group; as a result of the change in Glu and GABA levels, the Glu/GABA ratio increased significantly from 20 minutes and peaked at 120 minutes. Notably, this initial Glu reduction is consistent with the physiological role of dopamine in activating D2R, thereby inhibiting Glu release at the ends of the Glu cortical striatal pathway, while GABA reduction is associated with another physiological function of D2R, namely inhibiting the release of GABA from the axonal side branch of striatal projection neurons in the striatum.
Table 58 extracellular Glu and GABA levels in late PD striatum during DA signaling shutdown phase
Figure BDA0004007353830000641
L-DOPA: levodopa; PDSC1: a panaxadiol saponin composition;glu: glutamic acid; GABA: gamma-aminobutyric acid; glu/GABA: glutamic acid to gamma-aminobutyric acid ratio; ** p<0.01, *** p<0.001 (vs. control group); ### p<0.001 (vs. model group); n=5.
Late PD striatum in table 59 extracellular Glu and GABA levels during DA signaling on period
Figure BDA0004007353830000642
L-DOPA: levodopa; PDSC1: a panaxadiol saponin composition; glu: glutamic acid; GABA: gamma-aminobutyric acid; glu/GABA: glutamic acid to gamma-aminobutyric acid ratio; * p<0.05, *** p<0.001 (vs. control group); ### p<0.001 (vs. model group); n=5.
Glu and GABA changes in chronically treated L-DOPA animals are in marked contrast to changes in animals given the L-DOPA model at once. After the DA signal is turned "on", no early decrease in Glu is seen, but a rapid and sustained increase, indicating that chronic treatment with L-DOPA results in a decrease or loss of D2R inhibition of Glu release; glu levels reached a peak at 60 minutes, with peak rise times significantly shorter than 120 minutes for the model group and peak higher than for the model group), followed by a small slope drop and maintenance at a level significantly higher than for the model group (Table 59), with corresponding changes in area under the curve (Table 60). As can be seen, for middle and late PD, L-DOPA chronic treatment further exacerbates the rate, extent and maintenance time of elevated striatal Glu levels during dopamine signaling open periods, loss of D2R inhibition Glu release function results in rapid Glu elevation. In the L-DOPA chronic treatment group, animals had a decrease in GABA levels of 78.05% (significantly higher than 62.46% of the model group) at 20 minutes, 77.31% (significantly higher than 64.12% of the model group) at 60 minutes, 79.05% (significantly higher than 62.46% of the model group) at 80 minutes, and 78.55% (significantly higher than 23.26% of the model group) at 120 minutes. Accordingly, the Glu/GABA ratio increased rapidly and continuously after the DA signal was turned on, and peak levels could still exist to 120 minutes, 6.13 times 0 minutes (off period), significantly higher than 1.88 times the model group. These study data demonstrate that in vivo GABA levels in the striatum of animals receiving chronic L-DOPA treatment were reduced more rapidly, more widely and longer than in untreated animals in the "on" state of DA, indicating that chronic L-DOPA treatment resulted in a hypersensitive state in which D2R inhibited GABA release. The literature reports that the function of D2R in inhibiting GABA release from axonal side branches of projection neurons was found to become hypersensitive in PD models using electrophysiological characteristics [ Taverna S, et al, JNeurosci,2008;28 5504-5512, chronic L-DOPA treatment further enhances this hypersensitivity [ Wei W, et al, JNEurophylliosiol, 2016;117 (3):987-999]. Here, the applicant's research results functionally confirm that the chronic treatment of L-DOPA greatly aggravates the hypersensitivity phenomenon with the inhibition of GABA release function by PD striatum D2R, resulting in strong oscillation of GABA levels between DA off-period and on-period of the chronic treatment of L-DOPA in a range well above and below normal level. Based on the important physiological activity of GABA, this intense oscillation is sufficient in itself to trigger the corresponding pathological activity and to transform into pathological behaviour and symptoms.
Area under extracellular Glu and GABA level curves for late PD striatum during DA Signal on in Table 60
Figure BDA0004007353830000651
L-DOPA: levodopa; PDSC1: a panaxadiol saponin composition; glu: glutamic acid; GABA: gamma-aminobutyric acid, glu/GABA: glutamic acid to gamma-aminobutyric acid ratio; *** p<0.001 (vs. control group); # p<0.05, ### p<0.001 (vs. model group).
The following points are described in summary of the above study data: 1. the striatum of untreated middle and late stage PD rats shows a disruption of the excitatory and inhibitory balance characterized by a significant increase in both Glu and GABA levels and Glu/GABA ratios, a neurochemical characteristic closely related to motor symptoms of bradykinesia, muscle tone and tremors seen in PD patients; 2. acute (one-time administration) L-DOPA administration opens up a further increase in the DA signal and greatly reduces GABA levels, further exacerbating the otherwise present excitatory and inhibitory imbalance, it being apparent that the cumulative effects of this imbalance tend to lead to excitatory damage to striatal nerve structures (including various nerve endings and synaptic connections) and interneurons, exacerbating disease progression and progressively diminishing therapeutic effects for chronic therapies and inducing catabolism and its neuropsychiatric disorders in preparation for neurochemical conditions; l-DOPA chronic treatment further exacerbates the functional hypersensitivity of PD striatum D2R to inhibit Glu release and D2R to inhibit GABA release, thus further exacerbating Glu level elevation and corresponding GABA level changes during the "off" and "on" phases, resulting in a high Glu and Glu/GABA ratio that fluctuates dramatically up and down with "on" and "off" at a high level, while GABA oscillates between well above and below normal levels, respectively. As can be seen, the results of the study revealed a progressive decrease in the efficacy of chronic L-DOPA treatment, an end-of-dose effect, a progressive progression of the disease accompanying the treatment, and a neurochemical basis for its triggering of catabolism and neuropsychiatric disorders. Therefore, protecting or maintaining and reconstructing the neurochemical microenvironment in the PD striatum has important significance for treating PD and preventing and treating the weakening of the drug effect and toxic and side effects of L-DOPA treatment along with the prolonged treatment time.
It is particularly pointed out here that in Parkinson's Disease (PD) patients, in vivo DA deficiency caused by dopamine neuron degeneration leads to three motor symptoms (dyskinesia, tonic and resting tremors) and non-motor symptoms (NMS), including cognitive deficit, depression, apathy and anxiety. However, DA replacement therapies derived from L-DOPA overdose the DA receptor also result in a range of side effects including disabling catabolism, schizophrenic symptoms (e.g., hallucinations, delusions), dysregulated syndrome or impulse control disorders (ICDs such as hyperexcitability, pathological gambling and shopping, binge eating and excessive libido), notch action (i.e., abnormally repeated non-targeted behavior), and compulsive drug use. Our findings indicate that abnormal elevated extracellular Glu and GABA levels in the striatum are associated with PD movement and non-movement symptoms, and that disorders of striatal D2R function due to chronic treatment with L-DOPA and neurotransmitter disorders characterized by elevated Glu and reduced GABA are closely related to their neuropsychiatric side effects.
Study results 2: chronic administration of PDSC1 may reestablish middle and late PD striatal Glu and GABA homeostasis and combat secondary hits of PD neurochemical homeostasis by L-DOPA treatment.
PDSC1 or PDSC1 in combination with L-DOPA can repair Glu and GABA microenvironment in the striatum of middle and late PD rats and completely combat the destructive effects of chronic L-DOPA treatment. During the "off" phase, post 3 weeks of PDSC1 or pdsc1+l-DOPA treatment (at which time the course of the disease is 5 weeks), intra-striatal Glu and GABA levels were near normal animal levels (table 58). Since the extracellular Glu and GABA levels of the intrastriatum have deviated from normal levels four weeks after 6-OHDA injury, our results indicate that chronic PDSC1 administration restored the extracellular Glu and GABA balance of the striatum of PD and completely prevented the second disruption caused by chronic L-DOPA treatment. During the "on" period, the co-administered animals still maintained striatal extracellular Glu and GABA at normal levels, nor did the PDSC1 chronically administered animals see significant Glu and GABA level changes, but a small decrease in Glu occurred 120min after PDSC1 administration (Table 59 and Table 60). These data indicate that PDSC1 chronic treatment can completely block progressive disorders of PD striatal Glu and GABA levels, and can even restore Glu and GABA microenvironment within PD striatal; in particular, PDSC1 in combination with L-DOPA treatment not only completely prevents L-DOPA from interfering with Glu and GABA levels in the striatum, but also maintains the effect of PDSC1, suggesting that PDSC1 may protect the stability of the D1R function and the heterogeneous physiological function of D2R from non-physiological overstimulation by L-DOPA substitution. Therefore, it is expected that a combination of a superior PDSC composition with L-DOPA may reconstruct striatal neurochemical homeostasis, including DA, glu, GABA.
Accordingly, it is expected that PDSC-optimal compositions alone or in combination with L-DOPA may produce breakthrough in a variety of therapeutic effects, including systematic treatment of motor and other neuropsychiatric disorders and slowing or even blocking disease progression, slowing or even completely controlling the end-effects of L-DOPA therapy and its side effects, including catastrophe, neuropsychiatric disorders (e.g., hallucinations, irritability), cognitive decline or even dementia, impulse control disorders (e.g., pathological gambling and shopping, binge eating and superloving), notch behavior (i.e., abnormally repeated non-targeted behavior), and compulsive drug use, among others.
It is also specifically noted herein that striatal GABA-inhibitory and DA-signaling deficits are closely related to obsessive-compulsive disorder (OCD), attention Deficit Hyperactivity Disorder (ADHD) and Impulse Control Disorder (ICD), and Tourette's syndrome (TS, a hereditary neuropsychiatric disorder, manifested as a series of motor and non-motor symptoms during childhood, the major features of which are transient, notch, motor or vocal behavior, known as tics). Therefore, the results of the studies herein also support the medical use of the superior PDSC compositions for the treatment of these neuropsychiatric disorders.
Experimental results 3: PDSC1 is unable to reverse the substantia nigra dense dopamine nerve loss of middle and late stage PD but in combination with L-DOPA can slow or even block progressive degeneration.
To clarify the above neurotransmitter findings independent of the state of health of the nigrostriatal dopamine neuropathway, model and treatment animals were analyzed for substantia nigra dense part dopamine neurosurvival. The results of TH immunohistochemical analysis showed that about 98% of the dopamine neurons in the dense area of the black matrix had been lost in the PD model group (at this time the course was 5 weeks), and that the same severe nerve loss was also present in the L-DOPA group and the PDSC1 group, but 12.25% of the dopamine neurons survived in the combination group, and this data was significantly higher than in the other groups. These data demonstrate that PDSC1 and its combination L-DOPA have no restorative effect on the degeneration of the already existing substantia nigra neurons in the middle and late stages, but the combination of both can slow or even block further degeneration. The action of PDSC1 against elevated Glu levels of L-DOPA supports this neuroprotection, which further reveals that PDSC1 combined with L-DOPA treatment has a characteristic efficacy that slows or even blocks PD disease progression.
However, about 70% of dopamine nerves are known to be lost to motor symptoms, so the degree of slowing down the neurodegenerative effects produced by combination therapy is insufficient to significantly alleviate PD motor symptoms and other neuropsychiatric disorders. Moreover, the effect of PDSC1 in regulating Glu and GABA level homeostasis in the PD striatum is independent of the state of health of the nigrostriatal dopamine pathway. This further demonstrates the therapeutic value of the optimal composition for patients with middle and advanced PD.
Summary and discussion
PDSC1 can maintain or even reconstruct Glu and GABA neurochemical homeostasis in PD striatum. It is also noted here that, due to technical limitations that the current failure to measure the acetylcholine (Ach) level of the striatum, there are two cues that PDSC1 could also regulate PD acetylcholine homeostasis. Firstly, the dopaminergic up-going system of substantia nigra can down-regulate the levels of Ach and GABA in the striatum, and consistently we observe that the GABA level in the striatum of the middle and late PD rats is increased and the GABA level is drastically reduced by switching on DA signals with L-DOPA, and the effect of correcting the GABA increase caused by DA deficiency and the GABA is drastically reduced by L-DOPA treatment. It is therefore reasonable to predict that PDSC1 has a similar regulatory effect on striatal Ach homeostasis. Secondly, the hyperactivity of signal caused by the excessive release of Ach is the neurochemical basis for the induction of stiffness in mice by the dopamine 2 receptor (D2 receptor) inhibitor Haloperidol (HAL), in which GABA excessive release may also be involved, it being evident that the powerful actions of PDSC1 and other optimal compositions against HAL induced stiffness predict their actions against HAL upsetting Ach and GABA levels in the striatum. In particular, the combination with L-DOPA prevents the L-DOPA from disturbing the striatal nerve chemical homeostasis and the heterogeneous dopamine receptor functional homeostasis, and the combination of the two can slow or even block the further degeneration of the striatal dopamine nerve pathway. Therefore, it is expected that a combination of a superior PDSC composition with L-DOPA may reconstruct striatal neurochemical homeostasis including DA, glu and GABA. Central neurotransmitters are important components of the implementation of brain function signaling molecules, adaptation to changes in internal and external environments by down-regulation or up-regulation is an adaptive homeostasis, neurotransmitter disorders (elevated or reduced transmitter levels associated with stress or stress damage and no return to physiological levels after withdrawal of the stressor) are closely related to neuropsychiatric disorders, and also to side effects caused by drug therapy, in particular by drug therapy in the nervous system. As can be seen, the results of the above studies reflect the enhancement of PDSC1 found in example 12 with ATP, NAD + 、NAD + NADH ratio, NADPH andGSH is the effect of cellular adaptive homeostasis of signaling molecules, and further points out that PDSC1 can repair the effect of PD striatal cellular adaptive homeostasis even in the face of deleterious stressors (i.e., long-term exposure to L-DOPA), further supporting the medical use of PDSC1 and other potent ginsenoside compositions to slow down central side effects caused by drug therapy. In the following examples, we will further verify this medical use.
Further, our findings also predicted the medical use of PDSC1 and other optimal compositions in several ways.
First, PDSC1 and other superior ginsenoside compositions, particularly in combination with L-DOPA, are of great pharmaceutical value in the treatment of middle and late PD.
Parkinson's Disease (PD) is characterized by motor, cognitive, behavioral and autonomic symptoms, and treatment of PD with PDSC-optimal compositions alone or in combination with L-DOPA may produce breakthrough in a number of therapeutic effects, including systemic treatment of motor and other neuropsychiatric disorders and slowing or even blocking disease progression, slowing or even completely controlling the end-effects of L-DOPA treatment and its side effects, including catabolism and neuropsychiatric disorders (e.g., hallucinations, irritability), cognitive decline or even dementia, impulse control disorders (e.g., pathological gambling and shopping, binge eating and excessive sexual desire), flinching behavior (i.e., abnormally repeated non-target oriented behavior), and compulsive drug use.
Second, PDSC1 and other superior ginsenoside compositions treat a group of neuropsychiatric dysfunctional disorders characterized by striatal Glu hyperexcitability, insufficient GABA inhibitory signals, and DA signal hyperness.
Basal Ganglia (BG) is a group of nuclear clusters located under the cortex of the brain, connecting with the cortex, thalamus and brain stem. Its main functions include motor control, reinforcement learning, emotion motivation and other advanced cognitive functions. The striatum is the core of the basal ganglia, mainly receiving excitatory inputs with Glu as transmitter and DA inputs from the substantia nigra dense part dopamine nerves from the cerebral cortex and thalamus, while the local two intermediate neurons of the striatum take GABA and Ach as transmitters, respectively. The transmitter signals are mutually integrated and then output through spiny neurons (SPNs) to realize the regulation and control on cognition, emotion, motor function and habitual behavior. Therefore, striatal dysfunction is involved in various neuropsychiatric diseases such as PD, schizophrenia and depression. More and more studies have demonstrated that the neural activity of the different functional regions of BG is mainly a result of the balance between Glu excitatory neurons and gabaergic inhibitory neurons, and is also strongly influenced by the dopaminergic and 5-hydroxytryptamine energy systems. Consistently, insufficient striatal GABA inhibitory signals and hyperactivity of DA signals, or in combination with concomitant dysfunction of the 5-hydroxytryptamine energy system, will lead to a range of mental dysfunction disorders including: positive symptoms of schizophrenia and symptoms of cognitive disorders (therapeutic agents are mainly dopamine 2 receptor inhibitors, including classical inhibitors haloperidol and the novel inhibitors clozapine, thiopride, sulpiride and risperidone, etc.), obsessive-compulsive disorders (OCD, therapeutic agents are mainly serotonin reuptake inhibitor antidepressants), impulse control disorders (ICD, antidepressants and anxiolytic drugs are mainly used), certain forms of anxiety disorders (such as phobia and panic) and depressive states, and a group of childhood neuropsychiatric disorders including attention deficit hyperactivity disorder/disorder (ADHD, the main treatments are tomoxetine and 5-hydroxytryptamine reuptake inhibitors SSRIs), autism (the main treatments include antidepressants, typical and atypical antipsychotics, sodium valproate and carbamazepine and other mood stabilizers, B6-represented nutritional substances that improve neurotransmitter generation, antagonize the morphine drug sodium drolone), tourette syndrome (GTS or TS, a hereditary neuropsychiatric disorder that manifests as a series of motor and non-motor symptoms in childhood, the main features of which are transient, notch, motor or vocal behavior, known as tics, the treatments are mainly dopamine 2 receptor inhibitors including sulpride, haloperidol, clonidine, risperidone, quetiapine, olanzapine and the like) and Tourette syndrome (Tourette, characterized by frequent motor and vocal cord twitches, the most commonly used drugs are haloperidol). In particular, PV-inhibiting interneuron (PV-Ins) dysfunction in the anterior striatum affects the different loops of the cortical-basal ganglia and leads to motor tics and behavioral disorders of the GTS. The PV-Ins deficiency of the prefrontal cortex and hippocampus is assumed to affect the mental disorder and positive symptoms of schizophrenia, respectively, and the insufficient release of GABA function by PV-Ins also results in the recurrence of the disease in schizophrenic patients susceptible to stress and negative mood.
It is also noted here that neuropathological and neuroimaging studies indicate that most neurodegenerative and neuropsychiatric diseases manifest as significant astrocyte atrophy and loss of function. Loss of atrophic astrocytes to maintain homeostatic function is thought to be the pathophysiological cause of schizophrenia, major Depressive Disorder (MDD) and bipolar disorder (BPD). Astrocytes are critical for maintaining extracellular Glu and GABA at normal levels, so astrocyte atrophy and loss of function must lead to increased excitability of Glu in the brain of patients with neurodegenerative and neuropsychiatric diseases, further exacerbating the imbalance between excitability and inhibitory effects caused by insufficient GABA function.
Thus, the actions of PDSC1 to protect astrocytes (examples 9 and 10) and PV interneurons against loss of mitochondrial complex I function (example 11) and to maintain and even reconstruct Glu and GABA neurochemical homeostasis in PD striatum (example 13) can treat the above neuropsychiatric diseases against disease mechanisms which are different from those of the existing related drugs, and it is therefore reasonable to expect that PDSC1 and other superior ginsenoside compositions alone or in combination with the existing respective therapeutic drugs or neurotrophic drugs (including mecobalamin, vitamins B1 and B6, coenzyme Q10 and GSH precursor amino acids) can provide a safer and more effective treatment for patients.
Specifically, PDSC1 and other optimal compositions may create new hopes for the patient in the following: 1. the medicine is a safer and more effective therapeutic medicine for patients suffering from Attention Deficit Hyperactivity Disorder (ADHD), autism, tourette syndrome (GTS) and Tourette syndrome, and can improve the efficacy and reduce the toxicity of western medicines when combined with the existing medicines due to the difference of disease-related neurotransmitters regulated and controlled by the existing medicines. The dopamine 2 receptor inhibition currently used for the treatment of autism, GTS and tourette's syndrome can cause extrapyramidal side effects and other central and peripheral side effects including insomnia or sleepiness, dreaminess, anxiety, dysphoria, headache, dizziness, hypodynamia, dry mouth, constipation and the like to varying degrees, and obviously these side effects are more harmful to patients in childhood. The effects of PDSC1 and other potent compositions against the dopamine 2 receptor inhibitor haloperidol induced stiffness in mice supports their synergistic attenuation of dopamine 2 receptor inhibition. In particular, it is believed that increasing central neural network integrity and adaptive homeostasis is a process and objective of brain maturation in children and adolescents. Therefore, the effect of PDSC1 in enhancing cell adaptive homeostasis (strengthening body resistance) might also promote the neurological development of these infants, thereby fundamentally treating this group of childhood neuropsychiatric disorders and promoting the mental development and improving learning ability of the infants. It can be seen that PDSC1 and other optimal compositions are of particular therapeutic interest for these infants. Treatment of schizophrenia with combination of a PDSC1 or other optimal composition with classical and novel dopamine 2 receptor inhibitors overcomes the inability of these inhibitors to ameliorate negative symptoms such as cognitive impairment and extrapyramidal side effects in patients and provides better efficacy in alleviating positive symptoms. In particular, the course of schizophrenia generally persists, with recurrent attacks, exacerbations or worsening, and some patients eventually experience decline and mental disability, with only a few patients remaining healed or substantially healed after drug therapy and psychotherapy. The stress vulnerability (i.e., the vulnerability to adaptive homeostasis) caused by inadequate PV-INs function is a significant cause of recurrent episodes of disease due to internal and external stressor stimuli. It can be seen that the protective effect of PDSC1 on PV-INs and the effect of enhancing cell adaptive homeostasis both support the medical use of PDSC1 and other superior ginsenoside compositions for maintenance therapy to prevent disease recurrence.
Third, the medical use of PDSC1 and other optimal compositions for the prevention and treatment of alcohol and drug addiction, juvenile gaming addiction, pathological gambling, and various compulsive behaviors.
Addiction is a major social problem, and no good solution exists until now. Drug addiction, including smoking, alcoholism and illegal drug use, indirectly or directly results in global annual production1180 ten thousand deaths, this number is higher than cancer deaths and accounts for one fifth of the total deaths according to global disease burden studies. Game addiction for primary and middle school students (teenagers) places a great burden on the physical and mental health and academia of the child. The deficiency of dopamine 2 receptor (D2 receptor) is known to lead to a high risk of individuals with a variety of addictive, impulsive and compulsive behavioral tendencies, such as severe alcoholism, cocaine, heroin, cannabis and nicotine use, pathological gambling, sexual addiction, ADHD, tourette's syndrome, autism, chronic violence, post-traumatic stress disorder, conduct disorder and antisocial behavior (J Psychoactive Drugs,2000nov,32s I IV, 1-112). Consistently, alcohol and almost all drugs are met by patients with increased dopamine release, however long-term DA overdose may further impair otherwise low D2R function and overall dopaminergic circuit changes, resulting in stronger drug dependence and intense withdrawal symptoms for the smoker/patient. In addition, insufficient GABA inhibitory function and Glu hyperexcitability directly mediate withdrawal symptoms. Thus, the drugs currently being used or being tested in clinical trials for addiction treatment include two classes of active substances: promoting GABA function and inhibiting Glu signaling, such as acamprosate (a non-specific GABA receptor agonist), topiramate (an anticonvulsant that inhibits presynaptic voltage-gated sodium and calcium channels thereby inhibiting Glu release, activates type A GABA receptors and inhibits glutamate AMPA receptors), lamotrigine (an antiepileptic that inhibits presynaptic voltage-gated sodium and calcium channels thereby inhibiting Glu release), gabapentin (an anticonvulsant that inhibits presynaptic voltage-gated Na + And Ca 2+ Channel reduced Glu release), memantine (a non-competitive antagonist of NMDA receptors against Glu excitability), and the like. In sharp contrast to the above-described principles of drug therapy, PDSC1 acts both directly against the D2 receptor inhibitor HAL and against chronic treatment of L-DOPA to hyperstimulate the D2 receptor resulting in some of its functions being desensitized and others being hypersensitive and to bring back to near physiological levels too high Glu and GABA levels and too low GABA levels in the striatum of PD. It can be seen that PDSC1 and other optimal compositions can be directed against craving tendencies, during drug intake and during drug intakeThe overall regulation of neurochemical abnormalities during withdrawal has the unique advantage of altering brain homeostasis back to normal, so that PDSC1 and other optimal compositions are expected to have better therapeutic effects on relief of withdrawal symptoms than existing drugs and prevention of addiction and relapse that existing drugs do not. In particular, the effect of PDSC1 against D2 receptor inhibitor HAL induced stiffness in mice predicts that PDSC1 can slow down the effects of D2 receptor deficiency induced toxicity in a dopamine-independent manner, against D2 receptor deficiency eliciting pathological responses and abnormal psychological and behavioral (including alcohol addiction and toxicity behavior, addiction games or networks, pathological gambling, sexual addiction, autism, post-traumatic stress disorder, conduct disorder and antisocial behavior) as described above; the action of PDSC1 on inhibiting Glu release by D2 receptor but GABA release by Zeng Min D2 receptor in PD brain and on maintaining and reestablishing Glu and GABA steady state are weakened by PDSC1 on anti-L-DOPA treatment, which proves that PDSC1 and other optimal compositions can correct or return to normal functions of D2 receptor deficiency, insufficient GABA inhibitory and Glu hyperexcitability especially in withdrawal stage of addicts, and also support the use of PDSC optimal compositions in combination with dopamine drugs including L-DOPA for preventing and treating addictive behavior and neuropsychiatric disorder of addicts. Moreover, the above results also support that the use of a PDSC-effective composition in combination with existing alternative drugs (e.g., methadone, naltrexone) and non-alternative drugs (e.g., clonidine) is more effective in relieving withdrawal symptoms caused by addictive substances.
Game addiction, particularly network and video game addiction, is another common important social problem, and game addiction of primary and secondary school students (teenagers) creates a great burden on physical and mental health and academia of children, and adults are trapped in the game addiction and cannot be self-pulled. Network and video game addiction is associated with psychological and social co-morbidity such as depression, attention deficit/hyperactivity disorder, alcoholism, anxiety and poor psycho-social support. Such as those with higher severity of hyperactivity, are more prone to symptoms of addiction to electronic games and their negative consequences. In adults, obsessive Compulsive Disorder (OCD) is associated with addiction to network and video games. Although the specific neurophysiologic pathology of gaming addiction is currently unknown, intravenous general anesthesia (IVGA) is undoubtedly part of the "reward deficiency syndrome" caused by negative down-regulation of dopamine following excessive release of dopamine due to abnormal neurotransmitter interactions in the limbic system. The strong response of gaming addiction to treatment suggests that gaming addiction and other behavioral (procedural) addiction are similar to those previously described. From the perspective of the psychotic co-morbid state of network video addiction (including attention deficit/hyperactivity disorder, alcoholism, anxiety, depression, obsessive-compulsive disorder), the neurophysiologic pathology of these psychotic disorders, as previously described, D2 receptor deficiency, GABA dysfunction and Glu hyperactivity, etc., are also closely related to gaming addiction. Further, various antidepressants, mood stabilizers, antipsychotics, glutamine-like drugs, N-methyl-d-aspartate (NMDA) receptor antagonists (inhibiting Glu hyperexcitability) and the like are effective to some extent, further demonstrating the neurophysiologic pathology of mental disorders associated with gaming addiction Cheng Zheng. Currently, the combination of psychotherapy and psychoeducation is the primary approach to solving juvenile gaming addiction, which is clearly not effective in breaking the physical basis of gaming addiction (neurophysiologic pathology). Therefore, the drug therapy has good application prospect for the auxiliary treatment of IVGA and the management of co-disease symptoms. It is particularly pointed out here that adolescents are still in the process of developing a cerebral neural network, and positive learning of the process body concept and learning performance will undoubtedly promote this process. Therefore, the side effects of the existing related drug treatments on teenagers are more prominent and the negative consequences are more serious, and the effect of the PDSC1 on strengthening the adaptive cell homeostasis (strengthening body resistance and consolidating constitution) is also likely to promote the neural network development maturation of people with addiction tendency and patients with addiction. As described above, the PDSC1 and other effective ginsenoside compositions can be used for systematically treating the neuropathological network of game addiction and are highly safe, so that the ginsenoside compositions have important application value in preventing and treating teenager game addiction.
In summary, the PDSC-effective composition is used for preventing and treating alcohol and drug addiction, teenagers' game and network addiction, pathological gambling, autism, chronic violence, post-traumatic stress disorder, conduct disorder and anti-social behavior, and can be used for intervening treatment from the three-stage circulation of addiction including consumption/cheering/poisoning, withdrawal and negative influence and craving stages thereof, thereby providing a novel medicament for patients.
Example 14. Treatment of advanced PD with the optimal composition may result in breakthrough efficacy.
To reveal the potential that treatment of intermediate-and-late stage PD can produce multiple breakthrough drug effects, alone and in particular in combination with L-DOPA, which converts to DA in the brain that can replace DA severely absent from the intermediate-and-late striatum, applicants induced intermediate-and-late stage PD rat models with 6-hydroxydopamine (6-OHDA), studied the effect of chronic treatment of PDSC1, L-DOPA and both in combination on PD motor and non-motor symptoms in contrast to the unresolved clinical problem, and the side effects of PDSC1 on anti-L-DOPA treatment, after which the improvement effect of PDSC1 on the hallucinations of PD patients was determined using an acute model.
Example 14-1. Pharmacodynamic characteristics of PDSC1 for treating middle and late stage PD motor symptoms-alleviating symptoms and promoting motor function repair (treating both principal and secondary aspect), combination with L-DOPA can produce better efficacy for treating both principal and secondary aspect.
Grouping and administration: A6-OHDA-induced middle and late PD rat model was prepared as described above, and PD rats received PDSC1 (20 mg/kg), metoprolol (23 mg L-DOPA+5.75mg benserazide/kg, 7 times the dose per kg of human being, L-DOPA) and PDSC1+L-DOPA, respectively, by gavage for 21 days, once daily. And simultaneously setting a model control group and a normal control group.
Determination of the extent of disease: the extent of PD progression after modeling, changes in DA receptor sensitivity or disease progression caused by chronic treatment with L-DOPA, and antagonism of progressive changes in PDSC1 were determined by amphetamine-induced rotation assays. The measurement scheme is that the systemic rotation behavior to the same side of the injury is recorded for a period of 20-60 minutes after 2mg/kg amphetamine is injected into the abdominal cavity of a rat, every 20 minutes, every turn within 1 minute of the last 1 minute is recorded, 3 times are counted, and the result analysis is included by the average value of the 3 times of counting. The model is that the dopamine channel of the single-sided substantia nigra striata is degenerated, and the more severe the degeneration is, the more times the degeneration is rotated.
Kinematic behavior determination: rat forelimb dyskinesia was determined by the forelimb stride test (FAS) and motor coordination was assessed by the RotarodTest (rotamadtest). The specific procedure was as in the previous Rotenone (ROT) -induced PD rat model (example 3-2).
Table 61 Effect of chronic treatment of L-DOPA and PDSC1 and their combination treatment on motor function in late PD rats measured 1 hour post-dose
Figure BDA0004007353830000691
L-DOPA: levodopa; PDSC1: panaxadiol saponin composition 1; *** p<0.001 (vs. control group); # p<0.05, ## p<0.01, ### p<0.001 (vs. model group), n=5.
Acute action (palliative) of the drug and observation time point of drug effect of slowing down disease progression and repairing function (palliative): as shown in table 61, the day before the start of administration at 15 days after molding, amphetamine-induced rotation behavior, forelimb motor ability, and balance ability were measured to obtain pre-administration data for uniform grouping; sequentially measuring amphetamine-induced rotation behavior, forelimb exercise capacity and balance capacity 1 hour after administration on the first day to obtain data for improving exercise symptoms by one-time administration; measurements were made at the corresponding times on days 7, 14 and 21, and pre-dose data were also measured at these 4 time points in order to reveal the effects of the prosthetic motor function produced by the long-term treatment of PDSC 1. To further determine the efficacy of PDSC1 in potentially restoring motor function in PD animals, the motor capacity and balance capacity of the forelimb before and after the last administration was measured one week after the last administration was discontinued.
Study results 1: PDSC1 blocks disease progression and prevents dopamine receptor hypersensitivity caused by L-DOPA treatment.
Amphetamine-induced rotation assays were performed 1 hour after dosing on days 1, 7, 14 and 21 and the results are shown in table 61: all PD rats respond equally to amphetamine on day 1, with progressively increasing sensitivity in mid-to late-stage PD model animals as the course of the disease increases, with further increases in sensitivity by L-DOPA treatment, whereas the sensitivity of PDSC1 alone and L-dopa+pdsc1 combination treated rats from day 7 to day 21. The above data indicate that PDSC1 can prevent PD progression and the occurrence of dopamine receptor hypersensitivity caused by L-DOPA treatment. It is specifically noted herein that recent others reported that conditional D2R knockout mice have an increased motor response to amphetamine, indicating that L-DOPA chronic treatment-induced hypersensitivity is associated with reduced D2R function. As can be seen, the results of the study herein reflect the findings of example 13 that PDSC1 can completely combat the desensitization of the D2 receptor to inhibit Glu release and the hypersensitivity phenomenon to inhibit GABA release caused by chronic treatment with L-DOPA.
Study results 2: PDSC1 can rapidly improve the balance ability, and the efficacy is continuously good along with the prolongation of the treatment time, while L-DOPA can rapidly improve the forelimb exercise ability, but cannot improve the balance ability, but chronic treatment can reduce the balance ability; the combination of the two can maintain the respective medicinal effect advantages.
As shown in table 61, on day 1, single dose L-DOPA significantly enhanced forelimb function, PDSC1 was more effective in improving motor coordination, and the improvement effect of the two measures in combination was further enhanced. In particular, PDSC1 gradually restores forelimb function with prolonged administration time, and coordination is close to normal state and is also faster. In the FAS and Rotarod trial on day 21, both the forelimb function and motor coordination of pdsc1+l-DOPA animals were close to normal animal levels, indicating that the combination could improve their efficacy. In contrast, chronic L-DOPA treatment has side effects that impair motor coordination. This result is consistent with clinical inefficiency of L-DOPA on unbalanced (fall) treatment and further suggests that unbalanced (fall) events in advanced PD patients may be associated with side effects of L-DOPA treatment.
Study result 3: the PDSC1 chronic treatment can effectively promote the recovery of the motor function and the balance capacity of the forelimbs of the PD rats; the combined use energy of the L-DOPA and the PDSC1 is recovered to the normal level; the reparative effect of the chronic treatment of L-DOPA is mainly shown in the initial stage of the treatment and the reparative degree is limited, and the chronic treatment of L-DOPA can aggravate the degradation of the balance function, which explains the phenomenon that some middle and late stage patients frequently fall down after taking the L-DOPA.
To determine the ameliorating effect on the disease, FAS tests were performed prior to administration during chronic treatment (multiple doses) when there was no or little acute effect of the drug. As shown in table 62, the forelimb ability of PD rats with PDSC1, L-DOPA, or a combination of both was at the same level on day 7, but significantly better than the PD model group, indicating that limb function was partially restored for each treatment group at this time. However, as the course of the disease extended, PDSC1 and the combination continued to restore forelimb locomotor ability and the limb function reached near normal levels in animals of the combination group on day 21, whereas animals of the L-DOPA group did not continue to be restored from day 7. These data indicate that PDSC1 is effective in promoting recovery of motor function of forelimbs of PD rats, and that the combination of L-DOPA and PDSC1 can be recovered to normal level, and that the drug effect of L-DOPA is mainly shown in the early stage of administration.
TABLE 62 Effect of chronic treatment of L-DOPA and PDSC1 and their combination treatment on forelimb motor function in late PD rats measured 24 hours after dosing
Figure BDA0004007353830000701
L-DOPA: levodopa; PDSC1: a panaxadiol saponin composition; *** p<0.001 (vs. control group); ## p<0.01, ### p<0.001 (vs. model group), n=5.
To further confirm the efficacy of PDSC1 treatment to repair motor function in PD animals, fas and Rotarod assays were performed before and 1 hour after 5 days of dosing (day 28), respectively. As shown in table 63, all rats dosed for a long period had significantly improved forelimb function and locomotor coordination prior to dosing (5 days post-dosing), but the animals in the combination dosing group had the best limb function, close to normal animal levels, followed by PDSC1 group, with the least L-DOPA effect. This further confirms the efficacy of PDSC1 in the treatment of restoring motor function in PD animals, and is better when combined with L-DOPA. The residence time of the L-DOPA animals on the rotor bars was significantly shorter than that of the model animals 1 hour after dosing, and it was seen that the 21 day treatment course of L-DOPA destroyed the balance of PD rats, which explains why patients in the middle and late stages of more than 7 years after dosing typically fall after dosing. The above results demonstrate that: (1) PDSC1, particularly in combination with L-DOPA, restores motor function in PD rats; (2) For PD rats receiving L-DOPA chronic treatment, the DA signal phase of L-DOPA can enhance the movement function of the forelimbs of the PD rats, but simultaneously the movement coordination capacity is also destroyed, and the PDSC1 and the combined administration can not only improve the movement capacity and coordination capacity of the forelimbs to be close to normal level in the DA signal phase, but also stably restore the two capacities to be in a basically normal state.
Table 63 Effect of chronic treatment on motor function in late PD rats with L-DOPA and PDSC1 and their combination treatment measured before and after drug administration on day 7 of drug withdrawal
Figure BDA0004007353830000711
L-DOPA: levodopa; PDSC1: a panaxadiol saponin composition; *** p<0.001 (vs. control group); ### p<0.001 (vs. model group), n=5.
In summary, our research results show that PDSC1 not only can inhibit the progress of PD and restore motor functions, especially can rapidly improve and gradually restore the balance ability of PD animals in middle and late stages after administration, but also PDSC1 in combination with L-DOPA not only overcomes the defect of the balance ability of L-DOPA injury but also can synergistically produce better efficacy.
Example 15 treatment of advanced PD with PDSC1 in combination with L-DOPA may overcome the fatal drawbacks (side effects) of L-DOPA.
Example 15-1 combination therapy of PDSC1 with L-DOPA late PD overcomes the catabolism of L-DOPA.
Catabolism is a disabling side effect of L-DOPA and most PD patients receiving L-DOPA treatment eventually experience catabolism. To date, amantadine is the only drug approved by the U.S. FDA to slow down the treatment of rejection of L-DOPA, but the drug does not prevent rejection, only slows down existing rejection, and has serious side effects. Although the mechanism of the abnormality caused by chronic treatment of L-DOPA is not completely understood, it is considered that the process of inducing abnormality by L-DOPA should be a process of gradually moving the striatal neurochemical homeostasis and functional homeostasis between different subtypes of dopamine receptors to a disorder, and abnormality occurs when the change reaches a certain level. Thus, the effect of PDSC1 against chronic treatment with L-DOPA to disrupt the homeostasis of striatal Glu and GABA and the heterogeneity of functional homeostasis of the D2 dopamine receptor subtype may confer a pharmacodynamic profile for the prevention and treatment of catabolism. Accordingly, two studies were designed to verify this hypothesis, namely, PDSC1 in combination with L-DOPA, which can cause a dose of catabolism, to observe its efficacy against catabolism; and secondly, in combination with low-dose L-DOPA, the efficacy of the L-DOPA against the tendency to produce the catabolism is observed, and a research result supporting the presumption is obtained.
Example 15-1a PDSC1 and L-DOPA combination therapy for intermediate and late PD can prevent and treat L-DOPA catabolism.
Grouping and administration: the 6-OHDA-induced middle and late PD rat model was used as described above, and PD rats received PDSC1 (20 mg/kg), metoprolol (23 mg L-DOPA+5.75mg benserazide/kg), equivalent to 7-fold of the once-per-kg dose of human, called L-DOPA, and PDSC1+L-DOPA, respectively, by gavage for 21 days, once daily. The catabolism was measured after dosing on day 1, day 7, day 14 and day 21, respectively.
TABLE 64PDSC1 combination L-DOPA treatment corrects L-DOPA-induced catabolism
Figure BDA0004007353830000712
Figure BDA0004007353830000721
L-DOPA: levodopa; PDSC1: a panaxadiol saponin composition; ** p<0.01, *** p<0.001(vs.L-DOPA);n=5。
catastrophe Test (AIMs Test): PD rats received literature evaluations of the grade of catabolism by two trained laboratory workers blinded to the experimental group. At the designed time point, the rats are singly placed into an observation cage immediately after receiving the gastric lavage L-DOPA, the abnormal behavior is observed within 30-180 minutes after the injection of the L-DOPA, the abnormal behavior is observed every 20 minutes, and the time of 20 minutes is observed every 20 minutes, and the maximum time is 4 minutes. Thus, 6 scores were taken together and the average of 6 scores was included in the analysis. The catabolism was scored by three subtypes: orolingual AIMs, mandibular opening and closing, facial twitches, and tongue protrusion), limb AIMs, axial AIMs, tension torsion of the contralateral neck and torso to the lesion side. The severity score for each type of transaction was 0-4. The judgment standard is as follows: 0. no transaction exists; 1. duration less than 29 seconds or half; 2. lasting for 30-59 seconds or a majority of the time; 3. for the whole 60 seconds, but interrupted by an external stimulus (tapping the observation cage); 4. for the whole 60 seconds and without interruption by an external stimulus. The three AIMS subtypes add to get the total AIMS score.
Study results: as shown in table 64, the L-DOPA chronic treatment group animals developed a degree of abnormality after the first dose was given, and the abnormality was aggravated with the increase of the number of administrations; the animals in the PDSC1 chronic treatment group and the PDSC1 combined L-DOPA (PDSC1+L-DOPA) group gradually disappeared with the increase of the administration times. The research result shows that the combination of PDSC1 and L-DOPA can prevent and treat the abnormal movement.
Example 15-1b treatment of advanced PD with PDSC1 in combination with L-DOPA can prevent L-DOPA from causing a predisposition to or burning abnormality.
TABLE 65PDSC1 in combination with L-DOPA to prevent L-DOPA induced catabolism
Figure BDA0004007353830000722
L-DOPA: levodopa; PDSC1: a panaxadiol saponin composition; * p<0.05, ** p<0.01(vs.L-DOPA);n=5。
to further demonstrate that PDSC1 can completely prevent the onset of catabolism, chronic L-DOPA administration at lower doses induces a predisposition to catabolism, or ignition catabolism, followed by higher doses of L-DOPA to trigger catabolism. Animals received L-DOPA (12 mg/kg+3mg/kg benserazide) or PDSC1+L-DOPA for 21 days, respectively, to induce a predisposition to catabolism, followed by cessation of PDSC1 administration and a high dose of L-DOPA (23 mg/kg+5.75 benserazide) for 5 consecutive days to trigger catabolism. The results of the study are shown in Table 65, where L-DOPA chronically dosed animals developed a second trigger dose (day 2) followed by progressive exacerbation of the catabolism, whereas animals in the PDSC1+L-DOPA combination did not develop any catabolism after the first 3 trigger doses, but developed a significantly lighter intensity than the L-DOPA group, although at the last 2 trigger doses. It can be seen that PDSC1 in combination with L-DOPA can significantly combat the process of chronic treatment of burning signs.
The results of the two examples above are taken together to demonstrate that the combination of PDSC1 with L-DOPA not only prevents the onset of catastrophe, but also eliminates the pre-existing catastrophe. Amantadine is the only drug approved by the FDA for treating the abnormal condition clinically at present, but the drug only partially slows down the degree of the existing abnormal condition, cannot prevent the occurrence of the abnormal condition and has great toxic and side effects. It can be seen that the mode and intensity of action of PDSC1 against the adverse events is a breakthrough development compared to the existing therapeutic effects.
Example 15-2 PDSC1 prevents the decline in learning and memory due to chronic treatment with L-DOPA.
The potential effect of PDSC1 on L-DOPA-induced cognitive impairment in rats was determined using 6-OHDA-injured rats that did not meet the criteria for the mid-late PD model. For this purpose, the conventional Morris water maze method was used to examine whether PDSC1 could prevent L-DOPA from causing memory impairment. After Morris water maze training, the model animals were given L-DOPA (46 mg/kg, corresponding to the dose of the individual clinical treatment) or L-DOPA+PDSC1 (20 mg/kg) once daily for 28 consecutive days while the normal control group was set.
As shown in table 66, chronic L-DOPA treatment accelerated memory loss as represented by: on day 14, the L-DOPA group rats took longer to find the escape platform than normal animals. PDSC1 in combination with L-DOPA reduced memory impairment caused by L-DOPA and was observed to find escape platforms at day 28 at a time similar to normal rats.
TABLE 66 treatment of PDSC1 in combination with L-DOPA to significantly combat escape latency in the water maze in L-DOPA-extended rats
Figure BDA0004007353830000731
L-DOPA: levodopa; PDSC1: a panaxadiol saponin composition, which comprises a panaxadiol saponin, ** p<0.01, *** p<0.001 (vs. control group); # p<0.05(vs.L-DOPA);n=5。
example 15-3 PDSC1 prevents L-DOPA chronic treatment from causing mental performance impairment.
During the course of the example 15-2, it was unexpectedly found that the rats of the L-DOPA group developed abnormal aggression at week 3 of treatment, manifested as screaming and violent aggression, but did not develop any catastrophe, and then the aggression was continuously exacerbated, and each strike to the rearing cage by week 4 induced aggression of the animals within the cage, and these animals were also seen to be in a state of easy irritability. In clear contrast, the behavior of the rats in the group of PDSC1 combined with L-DOPA did not show any spontaneous aggression and irritability. The above observations suggest that PDSC1 can prevent mental disorders caused by chronic L-DOPA treatment.
It is specifically noted here that chronic (long-term) treatment of normal mice with high doses of L-DOPA is also known to induce aggression, and that its neurological basis is D2R-mediated nerve signal overactivation. Thus, the effect of PDSC1 in preventing aggression caused by chronic treatment with L-DOPA indicates that PDSC1 can maintain D2R stability against L-DOPA overactivation. This confirms from an important side that our study in example 13 found that chronic treatment of L-DOPA impaired the sensitivity of D2 receptors to inhibit Glu release, while increasing the sensitivity of D2 receptors to inhibit release of GABA by the spinous neuronal (SPN) axon side branch, resulting in a sharp increase in extracellular Glu of the striatum during the DA signal opening phase, a sharp decrease in GABA and an extremely disrupted Glu/GABA balance; the combination of PDSC1 with L-DOPA can maintain D2 receptor-mediated Glu and GABA release functions at normal levels, and it has been found herein that supported PDSC1 and other superior compositions treat PD and other neuropsychiatric disorders alone or in combination with clinically relevant first-line or commonly used drugs or agents that work in conjunction with each other.
Examples 15-4. Prediction of the medical use of PDSC1 in the prevention and treatment of schizophrenia such as delusional hallucinations in PD patients using an acute model.
Delusions induced by chronic treatment with L-DOPA are related to the function of D2Rs, so the effect of PDSC1 in combination with L-DOPA in maintaining dopamine receptor function homeostasis supports the medical use of PDSC1 in the prevention and treatment of delusions hallucinations in PD patients. The 5-hydroxytryptamine (5-HT 2A/2C) receptor agonist 2, 5-dimethoxy iodiamphetamine (DOI) induced head-swing behavior is a common positive symptom model of schizophrenia for which the FDA approves the drug pimavanserin (5-HT 2A receptor inhibitor or inverse agonist) for the treatment of hallucinations and delusions in PD patients. Thus, this model was used to further predict the hallucination and delusions symptoms that occurred in PDSC1 treated patients.
The test method comprises the following steps: a model of positive symptoms of schizophrenia was established by one-time administration of DOI hydrochloride (5 mg/kg, ip) to male ICR adult mice weighing 30+ -5 g, which had been acclimatized in the laboratory for 6-7 days. Animals were evenly divided into normal control, DOI model, and 3 dose PDSC1 (PDSC 140mg/kg, PDSC160 mg/kg, PDSC180 mg/kg, oral) groups, while clozapine (10 mg/kg, oral) positive control groups were set. Wherein, the mice are immediately given DOI hydrochloride after both PDSC1 and clozapine treatment groups are pre-administered for 30 min. Each mouse was used only once, 10 mice per group. The mice were evaluated for the level of schizophrenia positive symptoms using the shaking incubation period (i.e., the time between the start of the DOI injection time and the time the mice were first shaken) and the number of shaking hands within 5 minutes.
TABLE 67 DOI-induced head-swing behavior of PDSC1 against 5-HT (2A/2C) receptor agonists
Figure BDA0004007353830000732
Figure BDA0004007353830000741
PDSC1: a panaxadiol saponin composition; *** p<0.001 (vs. control group); # p<0.05, ### p<0.001 (vs. DOI model group); n=5.
Study results: as shown in table 67, mice of the DOI schizophrenia model group exhibited positive symptoms of schizophrenia relative to the normal group: the shaking latent period is obviously reduced, and the shaking frequency is obviously increased within 5 minutes. After PDSC1 pre-dosing treatment, the animal shaking head latency of the 3 dose group is obviously increased, the shaking head times within 5 minutes are obviously reduced, and the dose dependency is presented, wherein the drug effect of PDSC1 (80 mg/kg) is optimal, the drug effect is equivalent to that of clozapine, and the shaking head behaviors of mice are almost reduced to the normal level. The research result further supports the medical application of the PDSC1 for preventing and treating the schizophrenia symptoms of PD patients accompanied by hallucinations, delusions and the like, and supports the medical application of the PDSC1 for treating the schizophrenia positive symptoms.
Example 16 study of the efficacy of the Excellent composition in treating stroke (cerebral apoplexy).
Stroke (cerebral apoplexy) is an acute episode of cerebrovascular disease, whether hemorrhagic stroke or infarct ischemia or ischemia reperfusion injury caused by subsequent recovery of blood flow, directly injures the cells of each member of the neurovascular unit. In particular, according to applicant's study in example 9, malignant cycling between lesion member cells further exacerbates the damage to neurovascular units. Mitochondrial dysfunction, oxidative stress injury, inflammatory response are closely related to acute stroke and chronic brain injury. Therefore, PDSC1 protects the members of the neurovascular unit against the effects of the mitochondrial complex enzyme I inhibitor rotenone on inducing cytotoxicity and its mechanism of action against oxidative stress injury, inflammatory response and mitochondrial dysfunction induced by insufficient mitochondrial complex enzyme I function by maintaining ATP homeostasis predicts that PDSC1 can also protect brain cells and brain function against stroke injury and promote rehabilitation after stroke. Here, the prediction was validated with a mongolian sandy mouse whole brain ischemia reperfusion injury model and an angiotensin ET-1 induced rat focal brain ischemia model.
Induction of mongolian gerbil whole brain ischemia model: adult male Mongolian gerbil mice (60-75 g) were subjected to global ischemia via temporary bilateral carotid artery occlusion (6 min) under pentobarbital sodium (45 mg/kg, ip) general anesthesia, and then reperfusion. Another group of animals received sham surgery. During the whole procedure, the animal body temperature was maintained at 37±0.5 ℃ until the animal was completely recovered from anesthesia.
ET-1 induced focal cerebral ischemia and functional prognostic assessment: adult male Sprague-Dawley rats weighing 300-350 g were anesthetized with sodium pentobarbital (45 mg/kg, ip) and then placed in a stereotactic frame. Body temperature was maintained at 37.+ -. 0.5 ℃ with a heating pad, the skull was exposed, and a burr hole was placed on the dorsal lateral parietal cortex above the right side of the skull for injection of vasoconstrictor endothelin-1 (ET-1), and ET-1 (120 p moles in 4. Mu.L saline) was applied to the middle cerebral artery (near the striatal branch, AP, +0.9mm; ML,5.0mm; DV, -8.0 mm) producing prolonged vasospasms and subsequent ischemic injury. These stereotactic coordinates were determined from literature reports and initial results, with the needle held in place for 5 minutes. The other group received 4 μl of physiological saline as a pseudoischemic group (n=3). It is known that after ET-1 administration, cerebral blood flow is reduced to 30-50% of control levels, and significant reduction of the cortex and striatum persists for 16 hours. This reduced blood flow results in a large area of infarction, including injury to the ipsilateral dorsal and lateral neocortex and striatum. The pattern of this ischemic injury is similar to that reported previously after permanent occlusion of the middle cerebral artery in rats.
Neurological abnormalities were measured within 5 hours after recovery from anesthesia, then once daily for 6 consecutive days. Immediately after recovery from anaesthesia, animals that were successfully induced for stroke were confirmed to be substantially awake 1-2 hours after ET-1 injection. Stroke is characterized by the fact that the forepaws grip or cannot extend the contralateral forelimb when the animal is suspended by the tail, or rotate counterclockwise when the animal is placed on the floor. Rats that did not show any behavioral changes were excluded from the analysis. Neurological deficit within 1-5 hours after ET-1 injection was graded as described above: 0. no defects were observed; 2. contralateral forelimb flexion when suspended with tail; 3. the front jaw grips, crosses to the opposite side or spirals. Subsequent neurological abnormalities are based on the detection of abnormal gestures. Each rat was hung by tail above the table and chest torsion and forelimb extension were scored as follows: 0. defect-free, normal (forelimbs try to contact the table surface); 1. slight (chest torsion after 5 seconds of suspension); 2. moderate (torsion chest immediately after suspension); 3. severe (forelimb grip).
Experimental design and dosing: firstly, using a Mongolian gerbil whole brain ischemia reperfusion injury model, taking damage and loss of vertebral neurons in a sea horse 1 region as indexes, examining the efficacy of an optimal effect composition PDSC1 which is respectively administered once 1 hour before and after ischemia and then continuously treated for 6 days every day, wherein the doses are respectively 20, 40 and 60mg/kg, and simultaneously setting a positive control drug glutamate receptor NMDA receptor inhibitor MK-801. Study data show that the optimal composition can obviously resist damage and loss of neurons in the hippocampus induced by cerebral ischemia reperfusion after and before 40 mg/kg and 60mg/kg of administration on the 7 th day and the 30 th day of ischemia, wherein the drug effect of a large dose is better than that of a medium dose, and the drug effect of the large dose is obviously better than that of a positive control drug.
For the whole brain ischemia reperfusion model, the test groups included 3 dose groups (20, 40 or 60mg/kg, ig) of the sham-operated control group, the model control group, the MK-801 (3 mg/kg, ig) positive control group and PDSC 1. The control group was given an equivalent volume of physiological saline, once 1 hour before ischemia and then once daily. The animals of each group are subjected to histological analysis on brain tissues at 24 hours, 3 days and 7 days after ischemia, and the research results can reflect the acute injury effect of the drug on ischemia reperfusion. After obtaining the optimal PDSC1 effective dose, another animal batch was subjected to histological analysis of brain tissue at day 3 and day 21 of ischemia, and the results of the study further revealed whether the PDSC1 composition delayed neuronal damage. To better simulate the clinical situation, a further batch of animals received PDSC1 composition treatment 1 hour after ischemia, once daily for 6 consecutive days.
For ET-1 induced focal ischemia in rats, only the most potent dose of PDSC1 was observed in the sandy murine model. Rats were injected with saline or PDSC1 hour prior to ET-1 administration, followed once daily for 6 consecutive days. Histological analysis of brain tissue was performed at day 7 after ET-1 injection.
Histological analysis: at the indicated time points, brain tissue was fixed by cardiac perfusion with 4% paraformaldehyde in phosphate buffered saline (0.1M, pH 7.4) under deep anesthesia (Ulatan 1.2 g/kg) and was coronally sectioned with an oscillating microtome for 50 μm (rats) or 40 μm (gerbils). For sand mice, dorsal hippocampal slices were collected, with segments of 1.6 to 2.8mm postbregma. Six serial sections were defined as one group, and five groups were collected from each brain. The first plate of each group was stained with cresyl violet (Nissl stain) to observe viable neurons; the second patch was stained with 0.0004% fjb to observe dying neurons. The hippocampus CA1 was photographed under a microscope and then on a computer screen, 250 μm long viable neurons (Nissl staining with obvious nuclei and cell membranes) and dying neurons from the central region of the more severely damaged side CA1 were counted. The number of animals per animal was averaged and then averaged over each experimental group.
The forebrain coronal sections of rats, including the cortex and striatum, were collected about 2.6mm to-0.4 mm posterior to the bregma. Out of every 10 sections, 3 sections were left for staining, for a total of 5-6 groups (15-18 sections) per animal. The first patch of each group was stained with cresyl violet to observe surviving neurons, and the second patch was immunoreactive stained with heat shock protein 72 (HSP 72). HSP72 positive staining indicates that the nerve is experiencing damaging stress, and is used herein as a marker of nerve damage. HSP72 staining has a clear border and therefore semi-quantitative analysis was performed using NIH Image J software. Measurements were made on 5 to 6 sections of each animal evenly distributed over the lesion. The lesion volume was estimated using the following formula: volume= (a) 1 +a 2 +a n ) N x d, where d is the first sheet (a 1 ) To the last sheet (a n ) Distance between (unit: millimeter), a) 1 、a 2 、a n Is the lesion area (unit: square millimeter) of the corresponding slice.
Statistical analysis of data all parameters of each group of animals, including the number of neurons surviving or degenerating the CA1 region of the gerbils, the functional outcome score, and the lesion volumes of the rat cortex and striatum were averaged. Data are expressed as mean ± SEM. Test data were analyzed using analysis of variance in combination with Bonferroni test. P <0.05 was considered a significant difference.
Study results 1: the PDSC1 composition has curative effect on transient whole brain ischemia reperfusion of a gerbil.
As shown in fig. 32, after 7 days of ischemia reperfusion, severe nerve damage occurred in the CA1 region of the hippocampus on one side of all animals (n=13) in the ischemia model control group (normal saline control group), which was manifested by few surviving neurons identified by Nissl staining, and a large number of dying neurons identified by FJB staining occurred, of which 11 animals (85%) had severe damage on both sides. Thus, the data on the more severely damaged side of each animal was used to compare the experimental groups.
The PDSC1 composition (once 1 hour before ischemia, once daily beginning the next day, for 6 consecutive days) produced significant neuroprotection in a dose-dependent manner (fig. 32). Compared to the ischemia model group, no significant protection was seen at the lowest dose (20 mg/kg ig, n=8) for the PDSC1 group, whereas the number of surviving neurons was significantly increased by p <0.01 and the number of denatured neurons was significantly reduced in the PDSC1 group at 40 or 60mg/kg dose (n=8-12). MK-801 (3 mg/kg, ig, n=5) also produced significant neuroprotection p < 0.05), however, animals were running and out of balance within 10 minutes after injection of MK-801, followed by sedation, while animals injected with PDSC1 did not have significant behavioral changes. The research result proves that the drug effect of the PDSC1 is obviously better than that of the anti-glutamate excitatory injury treatment implemented by MK-801 in one week of the cerebral ischemia acute stage treatment, and the PDSC1 is highly safe.
To exclude the possibility that the PDSC1 fraction only delayed neuronal death, additional post-ischemic time points, i.e., days 1, 3 and 21 post-ischemia (fig. 33), were added to compare the treatment of nerve damage in the observed model control group with the PDSC1 group at a dose of 60 mg/kg. Animals received physiological saline 1 hour prior to ischemia (model control), no neurons were lost on day 1 after ischemia, CA1 showed some neurons lost on day 3, but a large number of neurons lost on day 7 and approaching the maximum, and the number of surviving neurons on day 21 (n=13) was not significantly different from that on day 7. No significant neuronal loss occurred at each time point in the treatment group with PDSC1, and there was no difference in the number of surviving neurons on days 7 and 21 (n=7). These data indicate that administration of PDSC1 hour prior to ischemia followed by a continuous treatment once daily for 6 days produces a sustained neuroprotective effect.
Next, to better simulate the clinical situation, animals received PDSC1 treatment 1 hour after ischemia, once a day for 6 consecutive days. Experimental results (fig. 34) indicate that PDSC1 at a dose of 20mg/kg had no significant effect (n=8) compared to the ischemia model control group, whereas PDSC1 at a dose of 40mg/kg (n=11) or 60mg/kg (n=6) could significantly increase the number of surviving neurons and decrease the number of degenerating neurons (p < 0.05). MK-801 (3 mg/kg, n=5) also had significant neuroprotection (p < 0.05), but was less protective than high dose PDSC1. The research result shows that the PDSC1 can play a neuroprotective role when being inserted 1 hour after ischemia, and the efficacy strength is obviously superior to that of the anti-glutamate excitatory injury treatment implemented by MK-801.
Study results 2: treatment of focal cerebral ischemia in rats with PDSC 1.
1-2 hours after ET-1 injection, the behavioral indications of 16 strokes in 18 model control animals included the forepaws gripping or failing to extend the contralateral forelimb and rotating on the floor to the contralateral side of the injury when the animals were suspended by the tail, with a model success rate of 88.9%. In the next 2-5 hours, 7 of these 16 rats successfully induced stroke exhibited seizure activity (including facial and forelimb clonus and head swing), with a seizure rate of 43.8%. In the next 7 days, there was no significant improvement in neurological deficit. In addition, 2 rats died in the control group (one died 50 minutes after ET-1 and the other died 3 days after ET-1). In clear contrast, 6 of the 7 animals in the PDSC1 group at the dose of 60mg/kg showed clear signs of stroke immediately after waking from anesthesia, indicating a 85.7% success rate of stroke, with no significant difference from the model control group. There was no significant difference between the middle segment behavioral index scores (2.9±0.1, n=6) and the model control group (fig. 35), indicating that PDSC1 could not directly combat cerebral ischemia caused by vasoconstriction due to ET-1, or that the two groups of animals were suffering from acute ischemic injury to the same extent. However, animals in the PDSC1 treated group did not develop seizures and die. Furthermore, the first day of ischemia behavioural index was significantly improved (p < 0.05), and the degree of improvement was further increased (p < 0.01) on day 7 (fig. 35). It can be seen that PDSC1 completely eliminates seizures and death in animals within 24 hours of the period of acute ischemia and significantly restores acute ischemia-induced motor dysfunction within 7 days.
As shown in fig. 36, the results of the histological analysis are consistent with the results of the observation of the motor behavior of the nerve function. In the ischemia model control group, there was severe neuronal loss in striatum and cortex. In the neuronal deleted region, HSP72 is positively stained, which is evenly distributed in the tissue, rather than showing individual neurons, the HSP72 stained region reflects the neuronal lost region. PDSC1 significantly reduced the damaged area and neuronal damage of the striatum and cortex of the ischemic area compared to the ischemia model control group.
Conclusion and discussion
The research results show that the PDSC1 effective composition can prevent epileptic attacks in the acute phase of cerebral ischemia injury, quickly restore the motor function of the acute phase of ischemia reperfusion injury, reduce the death rate in the acute phase and protect neurons in the ischemic brain area. This strongly supports the therapeutic value of PDSC1 for both the acute and chronic convalescence phases of stroke (stroke).
The research result also supports the medical application of the PDSC1 in preventing and treating vascular dementia and neuropsychiatric disorder caused by chronic vascular diseases. Dementia associated with cerebrovascular factors, commonly referred to as vascular dementia (vascular dementia, VD), is mainly caused by ischemic stroke, hemorrhagic stroke and related cerebral vascular hypoperfusion, which cause ischemia and hypoxia of brain tissues, or by lesions of extracranial large blood vessels and hearts, which indirectly cause ischemia and hypoxia of brain tissues, and finally leads to overall decline of brain functions. Furthermore, according to the applicant's discovery of the self-destructive behavior of diseased endothelial cells and astrocytes on neurovascular units, neurovascular units are, of course, both victims and participants in the development and progression of vascular dementia. In addition, ischemia and hypoxia of brain tissue caused by cerebral infarction or other causes of vascular diseases of relevant brain regions can also cause symptoms such as headache, dizziness, eye strain, vexation, irritability, insomnia, dreaminess and the like. Therefore, the medical application of the PDSC1 for preventing and treating vascular dementia and neuropsychiatric disorder caused by chronic vascular diseases is strongly supported by the effect of the PDSC1 for protecting the nerve and brain functions against the ischemia reperfusion injury of the whole brain and the focal ischemia reperfusion injury of the forebrain and the effect of protecting cell types of each member of the neurovascular unit against the toxicity induced by the deficiency/absence of the mitochondrial complex enzyme I.
The research result also supports the medical application of the PDSC1 optimal composition in treating the hypoxic-ischemic neonatal encephalopathy. Hypoxia and ischemia are common causes of neonatal encephalopathy, and common symptoms include frequent occurrence of conditions such as back-lying eyes and neck, stiff sleep, no excessive foot, easy waking up, irritability, and even convulsion, syncope and the like, and the symptoms last for more than 7 days, and sequelae can be left even recovery. Anoxic ischemic encephalopathy remains the primary cause of neonatal seizures. Moreover, tics, syncope (seizure) are also an independent factor and, together with primary nerve damage caused by hypoxia and ischemia, lead to the cachexia of the following neurodevelopment and sequelae of the ill neonate. The existing antiepileptic drugs have no neuroprotection effect, and the therapeutic side effects of the antiepileptic drugs can also negatively influence the normal development of the brain, and even can induce the apoptosis of the developing brain. PDSC1 protects the young fragile neonate brain from both ischemia-to-brain injury and seizure avoidance/reduction, without the side effects of existing antiepileptic drugs. Therefore, the PDSC1 optimal composition can provide safer and more effective treatment for neonatal hypoxic ischemic encephalopathy and other encephalopathy patients.
Shock is a pathological process in which the blood perfusion quantity of the organism is insufficient, so that the microcirculation system is blocked, and the metabolic disorder and the function damage of cells are caused, therefore, the anti-ischemia injury effect of the PDSC1 and the pharmacological effect of maintaining the energy metabolism and the redox homeostasis also support the PDSC1 to be independent and the medical application of treating shock by combining with the common anti-shock drugs.
Example 17 efficacy study of the combination of the Excellent composition with lamotrigine efficacy study of the treatment of epilepsy and bipolar depression or bipolar disorder/efficacy study of the Excellent composition in the treatment of epilepsy and in the modulation of the central excitatory and inhibitory balance.
Lamotrigine (LTG) is a presynaptic voltage-gated sodium channel inhibitor for use in the prevention of focal and systemic seizures in epileptic patients, as well as monotherapy or adjunctive maintenance therapy of bipolar disorders. However, lamotrigine has a number of common side effects including dermatitis and potentially life threatening severe dermatitis, adverse reactions to extrapyramidal systems (such as the appearance of parkinsonism or chorea-like movements), psychotic disorders (such as irritation aggression, ataxia, anxiety, confusion, hallucinations). The combination of the PDSC1 and the L-DOPA produces excellent drug effect and overcomes the toxic and side effects of the L-DOPA, and the action principle of the LTG is different from that of the PDSC1, so that it is reasonable to speculate that the combination of the PDSC1 and the LTG can synergistically produce drug effect and overcome the toxic and side effects of the LTG. Accordingly, the following studies were conducted, and the expected results of the studies were obtained.
Example 17-1 pdsc1 induces seizure against GABA synthesis inhibitors with better combination with lamotrigine.
The effect of PDSC1, LTG and PDSC1 combined LTG on acute seizures was observed with a model of rat acute seizures induced by the GABA synthesis inhibitors isoniazid and thiocarbamide. Here, these two models also serve to examine whether PDSC1 has GABA-like effects, and the results of the study may be further understood to provide insight into the effects of PDSC-optimal compositions on central excitatory modulation. Adult male SD rats (200-230 g) received three doses (10, 20 and 40 mg/kg) of PDSC1 or LTG (20 mg/kg), respectively, and 50mg/kg of thiocarbamide or 300mg/kg of isoniazid were intraperitoneally injected 1 hour after administration, 10 rats per group. Seizure monitoring was then performed for 3 hours, recording seizure latency, seizure class, and seizure duration for each animal.
As shown in fig. 37, the PDSC1 has a remarkable antiepileptic effect on both of the above acute epileptic models, and is expressed as follows: delaying epileptic seizures, i.e., extending seizure latency, reducing the level of severity of epileptic behavior, reducing the chance of seizure, suggests that PDSC1 may exert GABA-like effects when GABA is deficient in the brain. Consistent with the mechanism by which LTG inhibits epileptic seizures by inhibiting the release of the excitatory neurotransmitter Glu, LTG has no apparent anti-epileptic efficacy. Importantly, LTG in combination with PDSC1 can greatly enhance the antiepileptic efficacy of PDSC1, highlighted by further reduction in epileptic behavioral severity and number of animals presenting with large seizures.
It is also noted herein that the effect of the PDSC 1-optimizing composition against the GABA synthesis inhibitors isoniazid and thiocarbamide induced acute seizure in rats functionally demonstrates that the optimizing composition can directly combat seizures caused by the GABA-inhibitory dysfunction, indicating that the optimizing composition has GABA-like central inhibitory activity. As a presynaptic voltage-gated sodium channel inhibitor, LTG inhibition of glutamate release is critical in inhibiting seizures, and thus has poor efficacy against seizures caused by GABA deficiency. It can be seen that PDSC1 in combination with LTG can synergistically produce a powerful antiepileptic seizure effect by elevating GABA function and inhibiting Glu release.
Example 17-2 PDSC1 in combination with LTG has excellent therapeutic efficacy for refractory seizures and can overcome seizure rebound after withdrawal of LTG.
The epileptic drugs to date are not capable of treating the disease itself for the purpose of controlling epileptic seizures, and the degree of epileptic seizures after withdrawal is even heavier than before treatment, and are commonly called epileptic seizure rebound. Also about 30% of patients suffer from refractory epilepsy and respond poorly to existing antiepileptic drug therapies, including lamotrigine. Here, it was examined whether PDSC1 alone (5 mg/kg) or combined lamotrigine (10 mg/kg) could effectively control such refractory epilepsy, and whether the efficacy of PDSC1 or combined therapy could be continued or epileptic rebound occurred after withdrawal by comparing seizure conditions during and within 5 days after withdrawal to determine whether the drug had a palliative effect on epilepsy or could only exert an immediate efficacy (acute palliative effect).
The pilocarpine-induced rat chronic spontaneous epilepsy model is internationally recognized refractory epilepsy model, SD rats (male, 190-220 g) are induced by pilocarpine to generate sea horse and cortex injury according to the conventional method by using the acute seizure persistence of the pilocarpine, and then animals repair the damage for 3 weeks by themselves; spontaneous seizure behavior was observed starting at week 4, and was observed continuously for 8 hours per day, 9 a in the morning from monday to friday: from 30 to 5:30 pm, the epileptic seizure class, the number of seizures of the corresponding class, and the duration of a single seizure were recorded until the animals developed stable spontaneous seizures. The stable seizure refers to the occurrence of grade 3 or more in animals with 2 or more seizures per week for 2 consecutive weeks.
The epileptic animals were grouped into 7 groups, and the treatment of the tested drugs and the observation of the anti-epileptic drug effect were carried out. The medicine is administrated by stomach irrigation according to the set dosage, once daily for 26 days. Meanwhile, a model control group was set. Continuous observations were made for 8 hours a day, 9 a morning from monday to friday: and observing the epileptic level and the seizure times of the epileptic level at the corresponding level from 30 to 5:30 PM. As shown in fig. 38, ltg was able to fully control seizures for 9 days following treatment, but drug resistance occurred from day 19: seizures occur in nearly half of the animals. In clear contrast, the combination treatment of PDSC1 and LTG completely controlled seizures on the day of administration, and no seizures occurred except on day 4 throughout the course of the subsequent observation, but no significant therapeutic effect was seen for PDSC1 alone. Therefore, the drug effect of LTG for treating refractory epilepsy is weakened along with the extension of the treatment time, namely drug resistance phenomenon appears, the combined treatment of PDSC1 and LTG not only greatly shortens the effective time, but also overcomes the drug resistance of LTG treatment, and the special advantages of quick response and no drug resistance of the combined treatment are displayed.
To further observe the difference between LTG treatment and combination treatment in seizure status after withdrawal, another batch of chronic epileptic model animals was tested. The test groups included a model control group, a PDSC1 (5 mg/kg) group, an LTG (10 mg/kg) group, and a PDSC 1-combined LTG group, and were continuously administered for 10 days, followed by 5 days of withdrawal, and seizure frequencies of 3 and above levels during administration and withdrawal were observed with 5 days as an observation unit. As shown in table 68, the efficacy of each treatment group was consistent with the above test results during the administration period; during withdrawal, the seizure frequency of LTG-treated groups exceeded 2-fold prior to treatment, and the mean seizure frequency (2.14) was still lower than the mean seizure frequency (4.00) of the model control in the combined treatment, despite seizure onset, with no trend of increasing in the frequency of seizures in PDSC1 group compared to the period of administration. It can be seen that the combination of PDSC1 and LTG not only can overcome the rebound of seizures caused by LTG withdrawal, but also can produce sustainable anti-epileptic drug effects, which shows that the combination therapy can produce a primary drug effect on epilepsy.
The research results show that the combination of the PDSC1 and the LTG can also synergistically treat refractory chronic spontaneous epilepsy, and has the advantages of quick response, no drug resistance, no drug withdrawal rebound and continuous drug effect.
TABLE 68 influence of PDSC1 in combination with LTG on seizure in rats with chronic epilepsy before, after and after withdrawal
Figure BDA0004007353830000771
LTG: lamotrigine; PDSC1: a panaxadiol saponin composition; * p<0.05, ** p<0.01, *** p<0.001vs. pre-treatment; n=5.
Example 17-3 PDSC1 corrects elevated levels of Glu in spontaneous epileptic rats brain, in combination with LTG also increases GABA levels and antagonizes the dramatic increase in Glu and GABA levels caused by LTG withdrawal/PDSC 1 corrects for spontaneous epileptic rats brain excitatory and inhibitory imbalances and responds to withdrawal of lamotrigine.
The excitatory hyperactivity of neurotransmitter Glu and insufficient GABA inhibitory signals are the direct cause of epileptic acute attacks, and Glu signals further hyperactivity, oxidative stress injury and inflammatory reaction caused by each epileptic attack can further exacerbate brain dysfunction or injury which exists originally, so that the symptoms continuously progress, and excitatory hyperactivity caused by signal imbalance between neurotransmitter Glu and GABA is the key of epileptic attacks, and the PDSC1 can protect PD striatal PV positive GABAergic nerves, maintain the levels of PD striatal Glu and GABA in a stable state and can resist the damage of L-DOPA chronic treatment on striatal Glu/GABA balance. Therefore, applicants speculate that PDSC1 in combination with LTG may synergistically regulate excitatory and inhibitory balance of epileptic brain regions, and may even reestablish inhibitory and excitatory balance states in epileptic brain, thereby producing synergistic antiepileptic effects and treating epilepsy from the root. To verify this hypothesis, the effect of PDSC1, LTG, and combinations thereof on spontaneous epileptic rat hippocampal CA3 neurotransmitter Glu and GABA levels during treatment and after withdrawal was studied using a conscious animal brain microdialysis method in combination with LC/MS comparison.
The pilocarpine-induced rat chronic spontaneous epilepsy model animals were respectively perfused with PDSC1 (5 mg/kg), LTG (10 mg/kg) and PDSC 1-combined LTG once daily, while setting a model control group and a normal control group. For 7 days of administration, conscious animals were subjected to cerebral microdialysis to determine the hippocampal CA3 region Glu and GABA levels in a seizure-free state during administration. After administration for 10 days, the administration was stopped for 5 days, and the microdialysis was performed again on the 6 th day of the administration to determine the levels of Glu and GABA after the administration. Neurotransmitter levels were measured in the absence of seizure status, basal status (Base) and seizure (seizures), respectively, for the model control group.
As shown in tables 69 and 70, the Glu levels were significantly higher in the basal status without seizures than in the normal animals, with Glu levels further increased significantly during seizures, while GABA levels in both status were not significantly altered from the normal controls. This suggests that Glu-mediated excitatory hyperactivity is the primary cause of spontaneous seizures, and thus correction of this excitatory deviation is critical in controlling seizures and disease progression. Consistently, LTG can reduce Glu levels to levels near normal animals but affect GABA levels during treatment, whereas animals are in epileptic status during withdrawal, with significantly elevated levels of Glu and GABA, particularly Glu levels even higher than those in epileptic seizure status in model control animals. It can be seen that LTG inhibits spontaneous seizures mainly by inhibiting Glu release, and that withdrawal is associated with abnormally elevated Glu levels in the epileptic brain. Consistent with expectations, GABA and Glu levels were close in the PDSC1 group and normal control group during treatment and withdrawal, indicating minimal relief from correction of epileptic hippocampal Glu hyperexcitability states with PDSC1 treatment; the PDSC1 combined LTG group not only has Glu level close to that of the normal group, but also has GABA level which tends to be higher than that of other groups, which indicates that chronic treatment of the two groups can correct the Glu hyperexcitability state of the epileptic hippocampus and simultaneously improve the inhibitory function of GABA. It can be seen that chronic treatment of PDSC1 or its combined LTG corrects hyperexcitability in the epileptic brain by elevating GABA and lowering Glu, thus reverting back to a physiological or near-physiological central inhibitory and excitatory homeostatic state, and that this effect is not dependent on the acute effects of the drug but rather drug treatment restores the neurological state leading to hyperexcitability.
TABLE 69 influence of LTG, PDSC1 and combination therapy of both on extracellular Glu levels of Crimab CA3 for chronic epilepsy
Figure BDA0004007353830000781
LTG: lamotrigine; PDSC1: panaxadiol saponin composition 1; * P<0.001vs. normal control group; ### p<base level of model group 0.001 vs; &&& p<epileptic seizure phase of model group 0.001 vs; n=5.
TABLE 70 influence of LTG, PDSC1 and combination therapy of both on extracellular GABA levels of chronic epileptic hippocampal CA3
Figure BDA0004007353830000782
/>
Figure BDA0004007353830000791
LTG: lamotrigine; PDSC1: panaxadiol saponin composition 1; * p<0.05, *** p<0.001vs. normal control group; # p<0.05, ### p<base level of model group 0.001 vs; n=5.
The results of the studies in examples 17-2 and 17-3 demonstrate that the combination of PDSC1 and LTG synergistically modulates the excitability and inhibitory balance of the epileptic brain region, thereby correcting the hyperexcitability of the epileptic brain region Glu and reestablishing a sustainable inhibitory and excitatory balance state, so that the combination of both synergistically produces excellent antiepileptic effects (faster onset, stronger efficacy, no drug resistance) and can be used for treating epilepsy from the root (no disease progression, no drug withdrawal).
Example 17-4 PDSC in combination with LTG overcomes the side effects of LTG induced dermatitis.
Patients taking Lamotrigine (LTG) have a skin rash incidence of up to 10% and in order to increase the incidence of LTG-induced dermatitis in mice, the applicant used low concentrations of formaldehyde to induce dermatitis in animals receiving LTG. Adult ICR male mice (20-24 g) received lavage PDSC1 (10 mg/kg), LTG (20 mg/kg), and a combination of both, once daily, for 2 weeks, and the dehairing treatment was started on the backs of the mice on day 8, and 5 microliters of 5% formaldehyde was applied to the backs of each mouse on day 10, 1 time daily, and 3 consecutive days. Then, the animals of each group were analyzed for dermatitis occurrence rate by continuous observation for 3 days, and a typical individual was photographed.
As shown in FIG. 39, 5% formaldehyde by itself did not induce dermatitis in normal animals, but failed to induce dermatitis in all LTG treated animals (10/10), but failed to induce dermatitis in animals receiving the combined treatment of PDSC1 and TLG. It can be seen that PDSC1 in combination with LTG can completely combat the LTG-induced dermatitis response in mice.
Example 17-1 to example 17-4 summary and discussion of experimental results
The efficacy of LTG against acute epileptic seizure induced by insufficient GABA level is limited, and although the efficacy of LTG against acute epileptic seizure of chronic spontaneous epileptic animals is stronger than that of a PDSC1 optimal composition, the efficacy of LTG gradually weakens with the treatment time, which reproduces two major dilemmas of treating epilepsy clinically: firstly, the symptoms and root causes are treated, and the seizure rebound occurs on the day of drug withdrawal, so long-term medicine is needed; and secondly, refractory epilepsy is resistant to the existing antiepileptic drugs including LTG. The effect of PDSC1 on resisting acute epileptic seizure caused by insufficient GABA level is remarkable, the treatment effect on chronic spontaneous epileptic seizure is enhanced along with the treatment time, and the effect can last for one week after stopping the drug, which reflects the improvement of the functional status of epileptic brain. In particular, LTG as a sodium and calcium channel inhibitor can significantly inhibit glutamate release and thus rapidly control Glu-hyperkinetic state in therapeutic principle, we also observed that it inhibits GABA release; PDSC1 can enhance the inhibitory function of GABA and reduce Glu excitability, and especially improve the functional status of epileptic brain, so that the combination of the two can produce excellent drug effect by virtue of dominant complementation and can overcome the side effect of LTG. Indeed, the combined effect of PDSC1 and LTG against acute seizures initiated by insufficient GABA levels is further enhanced, greatly accelerating the process of controlling refractory seizures and overcoming the drug resistance phenomena, as well as overcoming the side effects of LTG causing dermatitis. Therefore, the research result strongly supports the medical application of the PDSC optimal composition combined with LTG for treating epilepsy, especially refractory epilepsy, and the combination can be expected to solve the clinical problem which can not be solved by the existing antiepileptic drugs.
The research results also support the medical application of the PDSC1 and other optimal ginsenoside compositions and LTG in treating bipolar disorder and Glu hyperexcitability or injury which are obvious pathological characteristics (including cerebral injury in acute stage of stroke, injury of central and peripheral nerves in shock, and intermediate and late PD movement symptoms and abnormal symptoms caused by dopamine drug treatment). Voltage-gated sodium channels widely regulate physiological functions of the central and peripheral, so LTG exerts its pharmacological actions (including inhibition of seizures, relief of hyperexcitability, and antagonism of glutamate excitatory central toxicity, etc.) by inhibiting glutamate release, while also causing toxic side effects of the central and peripheral, neuropsychiatric disorders (such as challenge aggression, anxiety, confusion, hallucinations, ataxia), adverse reactions of extrapyramidal systems (such as parkinson-like or chorea-like actions), dermatitis, etc. The applicant further points out that the rebound Glu and GABA increases sharply after withdrawal of chronic treatment of LTG indicates that chronic treatment of LTG as a sodium channel inhibitor causes the body to produce an adaptive change against the effect of LTG, which gradually weakens the pharmacological effect of LTG in lowering Glu levels, and also causes a sharp increase in Glu and GABA levels at no or low LTG levels in the brain during the interval of administration and withdrawal of treatment, thus causing its antiepileptic effect to gradually fade with prolonged treatment time, i.e. drug resistance, and withdrawal occurs. Clearly, severe deviations in Glu and GABA levels from normal levels tend to interfere with normal neuropsychiatric behavior and even lead to LTG producing various central side effects, including the neuropsychiatric disorders and extrapyramidal adverse reactions mentioned above. Based on the complementarity of the mechanisms of action of both PDSC1 and LTG, there is a strong justification for the safe and effective treatment of bipolar disorders and those characterized by Glu hyperexcitability or injury in combination with LTG by PDSC1 and other optimal compositions. It is also noted herein that the efficacy of PDSC1 in enhancing LTG antiepileptic effects and against side effects thereof also supports the synergistic attenuation of PDSC1 and other optimal compositions against other calcium and sodium inhibitors, particularly gabapentin.
Example 18. Study of the efficacy of the Excellent composition in the prevention and treatment of systemic lupus erythematosus.
Systemic lupus erythematosus (systemic lupus erythematosus, SLE) is an autoimmune disease, is frequently generated by women in childbearing age, has hidden onset, clinically presents butterfly erythema, arthralgia, light sensitivity, fever and the like, and can effectively control respiratory, nerve and internal organs if not, even endanger the life of patients and have extremely bad long-term prognosis; in addition, lupus is affected in other areas such as skin, joints, heart and lung, spirit, and organs. SLE, an unknown disease, is a major hazard to public health. The first line drugs for treating SLE at present are mainly immunosuppressants such as mycophenolate mofetil, cyclophosphamide, glucocorticoid and the like. These drugs have greatly improved prognosis for SLE patients, but they lack selectivity and specificity for immune response in the body, and have many toxic and side effects for long-term use, including reduction of body resistance-induced infection, influence on reproductive system functions, inhibition of bone marrow hematopoietic function, increase of tumor incidence, etc. The traditional Chinese medicine for treating SLE has long history, emphasizes overall appearance and treatment based on syndrome differentiation, and has the advantages of good long-term curative effect, small side effect, capability of reducing toxic and side effects of western medicines and the like. The following describes experimental studies of the effect of the beneficial agent PDSC1 on systemic lupus erythematosus disease.
Animal model: the immune enhancer pristane (prine) induced model mice have similar disease conditions to human SLE in clinical, immunological and pathological aspects, and are the most commonly used modeling method for artificially inducing SLE at present. BALB/c female mice (8 weeks old) were selected and 0.5mL pristane was injected intraperitoneally. On the 60 th day of modeling, urine of mice is collected, urine protein level is detected by ELISA method, urine protein of the mice is more than or equal to 0.30g/L, and the mice are determined to be successfully modeled, and SLE model mice induced by pristane are obtained.
Grouping animals: the SLE model mice induced by the pristinan are uniformly divided into four groups of a model group, a mycophenolate mofetil positive drug control group (MMF, 100 mg/kg), a PDSC1 group (5 mg/kg) and a PDSC1+MMF group (5+100 mg/kg) by taking the urine protein level as an index, 8 animals are administrated by lavage each day, and the model animals are administrated with an equal volume of physiological saline for 60 days continuously.
And (3) observing the indexes: (1) This symptom is also seen in SLE model mice, where typical skin erythema is common in SLE patients. Butterfly-shaped erythema on the cheeks of SLE patients and erythema on the cheeks of SLE model mice; (2) Lupus nephritis is the most common clinical manifestation of SLE patients, urine protein levels are an important basis for pathological changes in the kidneys, and mouse urine protein levels are detected 1 time every 15 days.
The experimental results are as follows:
(1) As shown in fig. 40, the SLE model mice developed beard drop around 70 days of molding, developed mild butterfly erythema around 75 days of cheek, developed moderate butterfly erythema around 80 days of cheek, developed moderate butterfly erythema around 120 days of cheek, and did not develop severe erythema.
(2) The change in appearance of each group of animals within 60 days of the dosing treatment is shown in table 71 and fig. 41. The model group 7 mice showed mild butterfly erythema on day 15, moderate butterfly erythema on day 21, no further development of butterfly erythema on day 60, and a butterfly erythema appearance ratio of 7/8; the MMF group 7 mice have mild butterfly erythema at 58 days of administration, and rapidly develop moderate butterfly erythema at 60 days of administration, wherein the butterfly erythema occurrence ratio is 7/8; the PDSC1 group of 3 mice showed mild butterfly erythema at 56 days of treatment, developed moderate butterfly erythema at 60 days of treatment, and the butterfly erythema showed a ratio of 3/8; the pdsc1+mmf group mice showed no butterfly erythema within 60 days of treatment. Experimental results show that PDSC1 has the effect of remarkably inhibiting butterfly-shaped erythema on the cheek of a SLE model mouse, and PDSC1 and MMF combined use can completely inhibit butterfly-shaped erythema on the cheek of the SLE model mouse.
TABLE 71 treatment of the number and extent of butterfly erythema in cheek of mice of each group for 60 days of dosing
Figure BDA0004007353830000801
(3) As shown in table 72, urine protein levels were significantly reduced in mice in MMF, PDSC1, and pdsc1+mmf groups, 15 days of animal administration; urine protein levels of pdsc1+mmf group mice decreased most rapidly at 30 and 45 days of dosing; the urine protein levels of mice in the MMF, PDSC1 and pdsc1+mmf groups were further reduced at 60 days of administration, and the levels tended to be consistent, with the urine protein levels of mice in the PDSC1 group being the lowest, indicating that PDSC1 reduced urine protein levels better than those in the MMF and pdsc1+mmf groups at the later stages of treatment.
TABLE 72 treatment of mice urine protein levels (g/L, mean.+ -. SD) for each group for 60 days of administration
Figure BDA0004007353830000811
In addition, consistent with the most common side effects (inhibition of hematopoietic function) of Mycophenolate Mofetil (MMF), it was also found during the experiment that both MMF group and pdsc1+mmf group mice developed a phenomenon of inhibition of hematopoietic function such as blushing of the nails after 30 days of treatment, which persisted throughout the treatment period, but this side effect was significantly lighter in pdsc1+mmf group mice than in MMF group mice, while PDSC1 group mice remained ruddy in nails throughout the treatment period, indicating that PDSC1 had an effect against MMF inhibiting hematopoietic function.
The combination of the experimental results shows that the active panaxadiol saponin composition (APDSC) is used singly or in combination with MMF to treat Systemic Lupus Erythematosus (SLE) diseases. Can play a role in reducing MMF dosage and enhancing attenuation when combined with MMF.
Since the active panaxadiol saponin composition (APDSC) can resist the immunopotentiator pristane to induce the mice to produce systemic red spot wolf disease, the medical application of APDSC for preventing and treating other autoimmune diseases and allergic diseases is supported, and the diseases include but are not limited to: hyper-reactive (allergic) lesions (including common allergic rhinitis, allergic asthma, urticaria, allergic conjunctivitis) and autoimmune diseases (dermatomyositis, scleroderma, rheumatoid arthritis) and over-immunity, neuromuscular diseases (multiple sclerosis, myasthenia gravis, demyelinating diseases), digestive diseases (chronic nonspecific ulcerative colitis, chronic active hepatitis, pernicious anaemia, atrophic gastritis), endocrine diseases (primary adrenocortical atrophy and chronic thyroiditis, juvenile-type diabetes), urinary diseases (autoimmune glomerulonephritis, pulmonary hemorrhagic syndrome), and rejection after liver and kidney transplantation. The results of the study (examples 17-4) of adverse effects of PDSC1 on the dermatitis caused by lamotrigine, combined therewith, also support the medical use of APDSC alone or in combination with MMF for the treatment of various refractory skin disorders including psoriasis.
Example 19 Holographic Ginsenoside Composition (HGC) and panaxadiol saponin composition (PDSC) for preventing and treating Alzheimer's disease and delaying aging.
Alzheimer's Disease (AD), also known as Alzheimer's disease, is an age-related chronic degenerative disease of the central nervous system. According to the world Alzheimer's disease report statistics in 2018, there are 5 tens of millions of diagnosed cases worldwide, and it is expected that the year 2050 will exceed 1.5 hundred million, which will bring a great socioeconomic burden. Clinical manifestations of Alzheimer's disease are mainly progressive memory and cognitive dysfunction, hidden onset, long latency, diffuse or plaque-like abnormal deposition of extracellular beta-Amyloid (Abeta), and neurofibrillary tangles caused by tau protein hyperphosphorylation in nerve cells are the most prominent pathological features in the brains of AD patients. However, most treatments for aβ and tau protein phosphorylation have proven clinically ineffective. Currently, the commonly used AD therapeutic drugs in clinic are cholinesterase inhibitors, excitatory neurotransmitter antagonists and drugs for AD complications, such as antidepressants, anxiolytics and the like. Furthermore, there are a number of new drugs against different pathogenesis of AD in the clinical trial phase, but the results are not clear. Accordingly, there is increasing interest in developing effective therapies for the treatment of AD (senile dementia), in particular, herbal medicines of natural origin with a multi-target synergistic mechanism.
The fast aging mice (Senescent acceleratedprone, SAMP 8) are cultured by special screening, and have uniform genetic background and stable aging pathological characteristics. SAMP8 mice have a lifespan of about 10 to 12 months, with rapid aging and cognitive impairment and pathological changes in the brain, very similar to the progressive occurrence of clinical AD. Therefore, compared with transgenic mice aiming at single pathological characteristics such as APP/PS1, the SAMP8 mouse is more suitable as an animal model for researching sporadic AD, and the SAMP8 mouse is also suitable for observing the aging delaying effect of the tested drugs.
Grouping and administration of animals: male, 6 month old SAMP8 mice 54 (available from Experimental animal science, university of Beijing, department of medicine, license number: SCXK 2016-0010). In order to bring the mental level of the animals of each group to the same state before treatment, the number of training times required for each mouse to find the escape platform in the water maze was measured by the conventional Morris water maze method, and then the animals were uniformly grouped according to the number and the therapeutic intervention was started, as shown in Table 73.
The mannite sodium capsule (Jiujiu-Shanghai Green valley pharmaceutical Co., ltd.) is used as a positive control drug for treating AD, and the holographic ginsenoside composition HGC1 (GRg/GRe/GRb/GRc/GRb 3/GRd =0.60/2.13/2.17/1.00/1.27/2.12) and HGC4 (GRg/GRe/GRb 1/GRc/GRb3/GRd =0.28/0.66/0.76/1.00/0.68/0.52) are both effective compositions for treating PD (GRb/GRc/GRb 3/GRd =2.17/1.00/1.27/2.12) and PDSC4 (GRb/GRc/GRb/3/GRd =0.76/1.00/0.68/0.52), but the effective compositions have different relative efficacy rates for preventing and treating senile dementia, and delaying senile dementia due to the relative arrangement of the individual components. The water maze test was performed at 1 month, 2 months and 3 months of treatment, respectively, and the death of the animals at any time was recorded.
TABLE 73 Experimental grouping and administration
Figure BDA0004007353830000821
Detecting the index:
1. survival curve
As shown in table 74, nine-phase one (new drug for treating AD) failed to improve animal survival after 3 months of dosing compared to model control. In contrast, in the case of the holographic ginsenoside composition and the panaxadiol ginsenoside composition including HGC1, PDSC1, HGC4 and PDSC4, no death occurred except for one animal in HGC1 group, and it was found that the ginsenoside compositions significantly reduced the death rate of animals at a dose of 5mg/kg once daily. The research results show that both the Active Holographic Ginsenoside Composition (AHGC) and the active panaxadiol saponin composition (APDSC) can prolong the life of the SAMP8 presenility mice.
TABLE 74 survival of animals
Figure BDA0004007353830000822
The differences between groups were analyzed using rank sum test. P <0.05, < p <0.01 compared to model control; compared to one of the nine phases, #p <0.05.
Platform finding Rate of Morris Water maze first day
Morris water maze tests were performed during 1 month, 2 months and 3 months of treatment, the latency period for finding an escape platform in each group at the first 2 time points is not different, the measurement result of the 3 rd month is shown in Table 75, one group in nine periods is not different from the model control group, the rate for finding an escape platform on the first day of animals in the PDSC4 group is highest, wherein 9 animals have 8 escape success, but the escape success rate of other ginsenoside composition animals is not obviously different from the model control group. According to the experimental result and the experimental result of animal survival rate, the PDSC4 can prolong the service life of the SAMP8 and can also remarkably improve the learning and memory capacity of the SAMP8, so that the PDSC4 can be used as a preferable composition for delaying aging and preventing and treating senile dementia.
Table 75 platform finding rate for the first day
Figure BDA0004007353830000823
The differences between groups were analyzed using rank sum test. P <0.01 compared to model control.
In conclusion, the holographic ginsenoside compositions HGC1 and HGC4 and the panaxadiol saponin compositions PDSC1 and PDSC4 can obviously prolong the service life of the SAMP8 mice with natural premature senility, and the PDSC4 can also obviously improve the learning and memory ability of the mice. Since damage and functional impairment of neurovascular units including cerebrovascular endothelial cells, astrocytes and nerve cells, mitochondrial dysfunction is accompanied by insufficient Adenosine Triphosphate (ATP) energy, oxidative stress injury and neuroinflammation are common pathological features of various neurodegenerative diseases including Parkinson's Disease (PD) and Alzheimer's Disease (AD) and are closely related to disease occurrence and development, previous studies have demonstrated that AHGC and APDSC containing two functional units of "GRb1+ GRd" and "GRc + GRb3" can protect damage and functional impairment of cerebrovascular endothelial cells, astrocytes and nerve cells, maintain and protect mitochondrial function, maintain ATP of energy substance at a sufficient level, prevent oxidative stress injury, inhibit NF- κb pathway and adhesion factors and release of inflammatory factors. The results of the above-mentioned our related studies support the medical use of Active Holographic Ginsenoside Composition (AHGC) active panaxadiol saponin composition (APDSC) in preventing and treating Alzheimer's disease and delaying aging.
It is also noted herein that although aging and neurodegenerative diseases and different neurodegenerative diseasesWith an adaptive imbalance in homeostasis (reduced ability to combat internal and external environmental disturbances by harmful stimuli), mitochondrial dysfunction, NAD + And ATP insufficiency, oxidative stress and chronic inflammation, neurovascular dysfunction, PV-positive neural mediated central inhibitory insufficiency, but the cause and mechanism of action of these pathological events may be different, so that the respective products of AHGC and APDSC may have a ratio of GRg/GRe/GRb 1/GRc/GRb3/GRd and a ratio of GRb/GRc/GRb 3/GRd, respectively, suitable for themselves. In the case of Parkinson's Disease (PD), all AHGC and APDSC have very significant effects on PD, both principal and secondary aspects of disease, and it is further determined that PDSC1 has more significant effects on epilepsy and cerebral ischemic injury. Therefore, aiming at delaying senility and preventing and treating different neurodegenerative diseases, even different phenotypes of the same disease or different physique of patients, the content configuration of the active ginsenoside contained in each of the AHGC and the APDSC discovered by the patent can be scientifically and freely selected so as to exert the curative effect of the active ginsenoside serving the patients.
Example 20 panaxadiol saponin composition (PDSC 1) protects the intestinal flora steady state against rotenone toxicity.
The human gut is populated with a large number of bacterial populations, and the health status of the bacterial populations correlates with the health status of the host. More and more studies indicate that protecting gut flora homeostasis is important for maintaining mental health, slowing down neuropsychiatric and other related diseases, and delaying aging. From the mechanism of action of the panaxadiol saponin composition PDSC1 for protecting the adaptive steady state of the organism based on maintaining the metabolic steady state and redox balance, to the action of the PDSC1 for resisting the injury of different drugs, toxic substances and pathological states on the organism, the action of the PDSC1 and other active (optimal) compositions for protecting intestinal flora and the related medical and health care application thereof can be predicted. Intestinal flora disorders are known to be one of the features of Parkinson's Disease (PD), and the mitochondrial complex enzyme I inhibitor Rotenone (ROT) -induced PD rats all have an intestinal flora disorder characterized by PD, and therefore, the prediction of protection of the intestinal flora by PDSC1 and other active (optimal) compositions is validated here by the ROT-induced intestinal flora disorder of PD rats. For this reason, we observed the effect of PDSC1 on ROT-induced dysbacteriosis in rats first, and then observed the effect of disrupting gut microbiota on PDSC1 against the formation of ROT-induced PD rat model.
The research method comprises the following steps:
effect of pdsc1 on ROT-induced dysbacteriosis in rat intestinal tract. The same procedure as in example 3-2 was used to induce PD in the rat model by ROT: rotenone was injected subcutaneously in 25% increments every 5 days, twice a day, once each of the morning (8:00) and evening (20:00). The molding dose is 0.5mg/kg in days 1-5, the molding dose is increased to 0.625mg/kg in days 6-10, the molding dose is 0.75mg/kg in days 11-15, and the volume of each administration is 0.05mL/100g. Male SD rats weighing 280-300 g were divided into a normal control group, a rotenone model group and a PDSC1 group, each group having 7 animals. PDSC1 (40 mg/kg) was administered by gavage half an hour before each injection of rotenone, and the normal control group was subcutaneously injected with an equal volume of sunflower oil and an equal volume of normal saline. At the end of the experiment, the clinical behavior symptoms of PD of each group of animals are detected, and the feces of each group of animals are collected and sent to a commercial establishment to determine intestinal flora before the behavior experiment is finished.
2. Disruption of intestinal flora effects of PDSC1 on antagonism of ROT-induced PD rat model formation. Animals were first consumed water containing antibiotics (including 1g/L ampicillin, 1g/L neomycin, 1g/L metronidazole, and 0.5g/L vancomycin hydrochloride) for 4 weeks to deplete the intestinal flora. Then, the animals were classified into an antibiotic control group, an antibiotic+rotenone group and an antibiotic+pdsc1+rotenone group, each group of 7 animals. The method of ROT-induced PD and the administration of PDSC1 are the same as those described above.
Study results:
pdsc1 protects gut flora homeostasis against ROT-induced PD rat gut dysbacteriosis. The fecal bacteria of each group of rats were subjected to data analysis at the mycological level using the 16SrRNA sequencing method of the microorganism. As shown in table 76, ROT significantly reduced the level of the flora of the Lachnospiraceae (Lachnospiraceae) and Lactobacillaceae (Lactobacillaceae) compared to the normal control, whereas the level of the flora of the Lachnospiraceae and Lactobacillaceae animals of the pdsc1+rot group were the same as the level of the flora of the normal animals and tended to be mathematically higher than the normal animals. It is known that the intestinal trichomonad and lactobacillus flora of PD patients are significantly reduced compared to normal persons, both of these bacteria are involved in the synthesis of short chain fatty acids (such as propionic acid and butyric acid), and that short chain fatty acids, particularly butyric acid, have a protective effect on the center, so that their reduced levels are considered to be an important cause of exacerbation of PD in flora disorders. Therefore, the increased proportion of these two species of bacteria in the flora of PDSC1 can be regarded as another form of the action of improving the adaptive homeostasis of the organism (strengthening body resistance and consolidating constitution), and is beneficial to the organism against the occurrence and development of ROT-induced animal PD. Taken together, the results of the above studies demonstrate that PDSC1 not only completely protects the intestinal flora from the toxicity of ROT, but also moderately increases the proportion of beneficial flora in favor of combating the occurrence and development of ROT-induced PD in animals.
TABLE 76PDSC1 up-regulates the bacterial levels of the Trichosporoceae (Lachnospiraceae) and Lactobacillus (Lactobacilliaceae)
Figure BDA0004007353830000841
Data are expressed as mean ± Standard Error (SEM); p compared to normal group using one-way analysis of variance<0.05; compared with the Rotenone (ROT) model group, # p<0.05;n=7。
2. disruption of intestinal flora may eliminate the improvement effect of PDSC1 on rotenone-induced motor dysfunction in PD animals. As shown in table 77, PDSC1 significantly improved Rotenone (ROT) -induced PD-like motor dysfunction in rats by a rotarod test, a forelimb stride test, a forelimb lift test, and a dysfunction score test; however, for animals treated with antibiotics for 2 weeks, resulting in disruption of their intestinal flora, PDSC1 lost its efficacy against the development and progression of ROT-induced PD in the animals. It can be seen that healthy intestinal flora or healthy gut is critical for PDSC1 to exert its effect in controlling PD, which further supports that protecting gut flora homeostasis may be one of the important mechanisms by which PDSC1 induces the formation and development of rat PD against the mitochondrial complex enzyme I inhibitor ROT.
TABLE 77 antibiotic treatment can eliminate the improvement of rotenone induced PD motor function by PDSC1
Figure BDA0004007353830000842
Data are expressed as mean ± Standard Error (SEM); using one-way analysis of variance, p compared to normal group <0.001; compared with the Rotenone (ROT) model group, # p<0.05, ### p<0.001; in comparison with the pdsc1+ro group, &&& p<0.001;n=7。
conclusion and discussion
The above results of the study are summarized to illustrate the following 3 key problems: pdsc1 may protect the gut flora against the toxicity of Parkinson's Disease (PD) risk factor pesticides present in the environment or against mitochondrial dysfunction in vivo; 2. the existing intestinal flora deficiency is unfavorable for the PDSC1 and other active (excellent) ginsenoside compositions to exert the efficacy of treating PD; 3. protection of the intestinal flora is also a mechanism of action of other active (excellent) compositions for controlling the PD-induced development and progression of rotenone.
Intestinal flora disorders are a characteristic part of the pathological network of PD, and consistent gastrointestinal dysfunction is the most common non-motor symptom of PD. Notably, changes in intestinal microbiota, damage to the Intestinal Epithelial Barrier (IEB), intestinal inflammation, and neuroplastic rearrangement of the Enteric Nervous System (ENS) are thought to be involved in the pathophysiology of PD intestinal dysfunction. As can be seen, our findings further reveal novel mechanisms of PDSC1 and other active (excellent) ginsenoside compositions systems for treating PD networks and medical uses for treating complications of gastrointestinal dysfunction in PD patients. Since the existing serious intestinal flora deficiency is unfavorable for the PDSC1 to play a role in preventing and treating the occurrence and development of PD, and the intestinal epithelial barrier and the enteric nervous system of a PD patient are abnormal and can negatively influence the healthy homeostasis of the intestinal flora, the research result also supports the medical application of the PDSC1 and other active (optimal) ginsenoside compositions in combination with the intestinal flora transplantation for treating the PD.
The research result further supports the wide medical application of the PDSC1 and other active (excellent) ginsenoside compositions for preventing and treating the neurodegenerative diseases and the neuropsychiatric disorders from the viewpoint of protecting the stable state of intestinal flora. In addition to PD, other neurological diseases, such as Alzheimer's Disease (AD), huntington's Disease (HD), multiple Sclerosis (MS) and Amyotrophic Lateral Sclerosis (ALS), are commonly associated with functional gastrointestinal disorders. These gastrointestinal disorders may occur at various stages of neurodegenerative disease to the extent that they are currently considered to be a component of their clinical manifestations. More recently, disorders, including depression, autism, and psychotic disorders such as schizophrenia, have also been found to be associated with disorders of the intestinal microbiota. Furthermore, each of the above diseases has a characteristic of a disorder of its own flora. At the other extreme of life, aging is associated with a diminished diversity of microorganisms, whereas the health state during normal aging is associated with a diverse microbiota. Further, changes in intestinal microbiota, damage to the Intestinal Epithelial Barrier (IEB), intestinal inflammation, and neuroplastic rearrangement of the Enteric Nervous System (ENS) may be associated with the pathophysiology of PD intestinal disorders. Thus, protecting the damage to the intestinal epithelial barrier, intestinal inflammation and the enteric nervous system is beneficial in protecting the intestinal flora homeostasis. The role of PDSC1 in protecting neurovascular units and blood brain barriers and in inhibiting central inflammatory responses is predicted to protect the intestinal epithelial barrier and the enteric nervous system and in inhibiting intestinal inflammation. In conclusion, our research results further support the medical use of PDSC1 and other active (excellent) ginsenoside compositions for preventing and treating neurodegenerative and mental disorder diseases and delaying aging.
In particular, the effect of PDSC1 protection of the intestinal flora against ROT toxicity predicts the effect of PDSC1 and other active (optimal) ginsenoside compositions to protect the intestinal flora homeostasis under other pathological conditions and aging processes. Therefore, our findings also support the health care effect of PDSC1 and other active (excellent) ginsenoside compositions on populations with fragile and deregulated intestinal flora.
Example 21. Active Total Ginsenoside Composition (ATGC) is prepared by comprehensively utilizing ginseng, american ginseng and pseudo-ginseng of Panax.
According to 10 feeding schemes designed in example 4, 10 samples of 10 active ginsenoside compositions (ATGC 1-ATGC 10) were prepared (1) composition ATGC1 of (1) "American ginseng root total saponin 1 part+American ginseng stem leaf total saponin 2 part, (2)" composition ATGC5 of (1) ginseng root total saponin 1 part+American ginseng stem leaf total saponin 1 part+3 part of (3) "composition ATGC2 of (1) ginseng stem leaf total saponin 2 part+ginseng stem leaf total saponin 1 part+American ginseng stem leaf total saponin 1 part), (4)" composition ATGC4 of (2) ginseng root total saponin 2 part+ginseng stem leaf total saponin 1 part+American ginseng stem leaf total saponin 2 part+composition ATGC4 of (5) "composition ATGC5 of (1) ginseng root total saponin 1 part+American ginseng stem leaf total saponin 1 part+1 part of (6)" composition ATGC1 of (1) ginseng stem leaf total saponin 1 part+2 part+7 of (1) stem leaf total saponin 1 part of (1) and (2) American ginseng stem leaf total saponin 1 part+1 part of (1) total saponin 1 part of (2) stem leaf total saponin 1 part of (1) and (7) stem leaf total saponin 1 part of (1) ginseng stem leaf total saponin 1 part of (1) and 10) total saponin of (1) total saponin 1 part of (1) American ginseng stem leaf total saponin 1 part of (1) total saponin 1 part of (1) and 2) total saponin of (1 part of) total saponin of (1) total saponin of). The contents of 7 ginsenosides GRg1, GRe, GRb1, GRc, GRb2, GRb3 and GRd of the composition (ATGC 1 to ATGC 10) were measured by HPLC method, and the relative proportions of the contents were analyzed.
Table 78 Ten feeding schemes comprehensive utilization of active total ginsenoside composition (ATGC 1-ATGC 10) obtained by total saponins of Ginseng radix, radix Panacis Quinquefolii and Notoginseng radix, wherein the content of each saponin is (%, n=3)
Figure BDA0004007353830000851
The analysis results are shown in Table 78 and Table 79, and 10 active total ginsenoside compositions (ATGC 1-ATGC 10) prepared by 10 different feeding schemes all meet the following conditions: the total amount of panaxatriol saponins and panaxadiol saponins is more than 60%, wherein the content of each of four ginsenosides GRe, GRb1, GRc, GRb3 and GRd is more than 4.80% and less than 20.00%, but the condition that the content of each of the four ginsenosides is simultaneously more than 10.00% is excluded; while GRg and GRb are each less than 8.00% in content, ranging from 2.12 to 5.91% and from 3.58 to 7.96%, respectively.
Further, the content configuration of each ginsenoside component in each ATGC composition meets the following eight ratio requirements, and the composition comprises the following components: total content of panaxadiol saponins/total content of panaxatriol saponins (TPDS/TPTS) =1.87-4.40, GRe/GRg 1=2.31-4.41, GRb 1/gre=0.64-1.86, GRb1/GRd =0.79-2.08, GRb1/GRc =0.67-2.17, GRb1/GRb 3=0.82-2.76, GRc/GRb 3=0.79-2.11, and (GRb 1+ GRd)/(GRc + GRb 3) =0.66-1.92; furthermore, the ratio of seven kinds of Chinese medicinal materials GRg/GRe/GRb 1/GRc/GRb3/GRd is kept to be 0.60/2.13/2.17/1.00/1.27/2.12, 0.76/2.16/1.38/1.00/0.63/1.74, 0.37/0.86/1.00/1.00/0.47/0.64, 0.28/0.66/0.76/1.00/0.68/0.52, 0.39/0.91/0.75/1.00/0.56/0.66, 0.23/0.91/1.39/1.00/1.09/0.91 and 0.21/0.76/1.01/1.00/1.09/0.73 respectively, and the ratio of eight kinds of the ratio and GRg/1/GRe/GRb/GRc/GRb/GRd are used as the preferred mixing ratio of the active Ginsenoside Composition (GC) and the total saponin or the total saponin preparation product thereof according to the standard are prepared by mixing ratio of the total saponin composition.
Table 79 Active Total Ginsenoside Composition (ATGC) content configuration of individual ginsenoside components
Figure BDA0004007353830000861
Note that: TPDS: total content of panaxadiol saponins; TPTS: total content of panaxatriol saponins.
As can be seen from the results of the above examples, the present invention discloses a method for preparing an active ginsenoside composition by comprehensively utilizing raw materials of ginseng, ginseng stem and leaf, american ginseng stem and leaf and pseudo-ginseng of ginseng genus or total saponin raw materials extracted from the raw materials, and uses of the active ginsenoside composition in medicine and health care, and product quality control standards based on the specific uses thereof.
Ginsenoside has wide biological activity and pharmacological action, is the main active ingredient in the ginseng traditional Chinese medicinal materials and the total saponin raw materials thereof, however, the content difference of individual active saponin ingredients in the traditional Chinese medicinal materials and the total saponin raw materials is obvious and has complementary characteristics. The invention fully utilizes the characteristic that the content of each single active ginsenoside in the traditional Chinese medicine and the total saponin raw materials is complementary, and scientifically mixes and configures different traditional Chinese medicine or total saponin raw materials by taking specific application as a guide to prepare high-purity Active Holographic Ginsenoside Composition (AHGC), active ginseng diol saponin composition (APDSC) and Active Total Ginsenoside Composition (ATGC) which are used for preventing and treating different diseases and health care.
Each active composition of the AHGC and the ATGC contains seven active ingredients of ginsenoside GRg, GRe, GRb1, GRb, GRb3, GRc and GRd, and eight ratios [ GRe/GRg1, GRb1/GRe, GRb1/GRd, GRb1/GRc, GRb1/GRb3, GRc/GRb3, (GRb 1+ GRd)/(GRc + GRb 3) and total panaxadiol saponin/total panaxatriol saponin (TPDS/TPTS) ] are respectively provided with certain range requirements; the APDSC active compositions all contain five active ingredients of ginsenoside GRb1, GRb2, GRb3, GRc and GRd, and the five ratios (GRb 1/GRd, GRb1/GRc, GRb1/GRb3, GRc/GRb3 and (GRb 1+ GRd)/(GRc + GRb 3)) of the content configuration have certain range requirements, and the reasonable eight ratio or the five ratio is the substance of compatibility of the active compositions and is also the basis of the preferred quality standard of the active composition products of AHGC, APDSC and ATGC and the mixing configuration feeding proportion of traditional Chinese medicinal materials or total saponin raw materials used for preparing the active composition products.
The scientific mixing configuration of the invention is based on the reasonable eight-ratio or five-ratio content of each active ginsenoside in each active composition, fully and reasonably utilizes the unique biological activity and pharmacological action of each single ginsenoside and the mutual synergistic action of each ginsenoside, in particular the subtle synergistic action (inexhaustible) of (GRb 1+ GRd) and (GRc + GRb 3) functions, therefore, each active composition of AHGC, APDSC and ATGC has wide medical and health care applications, and comprises the following steps: medical use for the prevention and treatment of neurodegenerative diseases having common pathological features, parkinson's disease, alzheimer's disease, vascular dementia, huntington's disease, multiple sclerosis and amyotrophic lateral sclerosis; medical application for preventing and treating mitochondrial myopathy, mitochondrial encephalomyopathy, pellagra, stress diseases, peripheral neuropathy, neuropsychiatric dysfunction diseases, apoplexy (cerebral apoplexy), epilepsy, systemic lupus erythematosus diseases and aging delay; medical application for improving sub-health state; medical application for preventing and treating alcohol addiction, drug and drug addiction, teenager game addiction, pathological gambling addiction, sexual addiction and various compulsive behaviors; health care application for improving sub-health state; synergistic and reduced side effects health care uses in combination with drugs for treating parkinson's disease or drugs for treating epilepsy.
The foregoing is merely a preferred embodiment of the present invention and it should be noted that modifications and adaptations to those skilled in the art may be made without departing from the principles of the present invention, which are intended to be comprehended within the scope of the present invention.

Claims (15)

1. An active ginsenoside composition, which is characterized by comprising a functional unit 1 consisting of GRb and GRd and a functional unit 2 consisting of GRc and GRb 3;
the mass ratio of the functional unit 1 consisting of GRb and GRd to the functional unit 2 consisting of GRc and GRb is 0.66-1.92.
2. The active ginsenoside composition of claim 1, wherein the active ginsenoside composition is an active panaxadiol saponin composition;
in the active panaxadiol saponin composition, the mass ratio of GRb to GRd is 0.79 to 2.08, the mass ratio of GRb to GRc is 0.67 to 2.17, the mass ratio of GRb1 to GRb3 is 0.82 to 2.76, and the mass ratio of GRc to GRb3 is 0.79 to 2.11;
preferably, the mass ratios of GRb, GRc, GRb3 and GRd include at least one of the following, but are not limited to: 2.17:1.00:1.27:2.12, 1.38:1.00:0.63:1.74, 1.00:1.00:0.47:0.64, 0.76:1.00:0.68:0.52, 0.75:1.00:0.56:0.66, 1.39:1.00:1.09:0.91 and 1.01:1.00:1.09:0.73; and allows the ratio of the components of each group to be varied within 10%;
Further preferably, the active panaxadiol saponin composition further comprises GRb2;
the total mass percentage of the active panaxadiol saponin composition is more than 85% of GRb, GRc, GRb2, GRb3 and GRd, wherein the content of GRb2 is 0.1% -16%.
3. The active ginsenoside composition of claim 1, wherein the active ginsenoside composition is an active holographic ginsenoside composition or an active total ginsenoside composition;
the active holographic ginsenoside composition or active total ginsenoside composition comprises panaxatriol saponin and panaxadiol saponin; the panaxatriol saponins are mainly, but not limited to GRg and GRe; the panaxadiol saponins are mainly but not limited to GRb1, GRb3, GRc and GRd; wherein the ratio of the total mass of the panaxadiol saponins to the total mass of the panaxatriol saponins is 1.88-4.41, the mass ratio of GRe to GRg1 is 2.31-4.41, the mass ratio of GRb1 to GRe is 0.64-1.86, the mass ratio of GRb1 to GRd is 0.79-2.08, the mass ratio of GRb1 to GRc is 0.67-2.17, the mass ratio of GRb1 to GRb3 is 0.82-2.76, and the mass ratio of GRc to GRb3 is 0.79-2.11;
preferably, the mass ratios of GRg, GRe, GRb1, GRc, GRb3 and GRd include at least one of the following, but are not limited to: 0.60:2.13:2.17:1.00:1.27:2.12, 0.76:2.16:1.38:1.00:0.63:1.74, 0.37:0.86:1.00:1.00:0.47:0.64, 0.28:0.66:0.76:1.00:0.68:0.52, 0.39:0.91:0.75:1.00:0.56:0.66, 0.23:0.91:1.39:1.00:1.09:0.91 and 0.21:0.76:1.01:1.00:1.09:0.73; and allows the ratio of the components of each group to be varied within 10%;
Further preferably, the active holographic ginsenoside composition or active total ginsenoside composition further comprises GRb;
still more preferably, the total mass percentage of the active holographic ginsenoside composition is more than 70% of GRg, GRe, GRb1, GRc, GRb2, GRb and GRd, based on 100%, specifically comprising the following components in percentage: 3.22 to 7.71 percent of GRG1, 11.99 to 21.87 percent of GRe,12.62 to 19.82 percent of GRb1,8.42 to 18.82 percent of GRc,5.22 to 10.45 percent of GRb2,6.35 to 17.14 percent of GRb3 and 9.83 to 17.85 percent of GRd, but the condition that the sum of the content upper limits of the components is equal to or more than 100 percent is not included;
the total mass percent of the active total ginsenoside composition is more than 50 percent, calculated as 100 percent, of GRg1, GRe, GRb1, GRc, GRb2, GRb3 and GRd, and the active total ginsenoside composition specifically comprises the following components in percentage by weight: 2.12 to 5.91 percent of GRG1,9.05 to 16.77 percent of GRe,9.31 to 19.12 percent of GRb1,6.42 to 14.33 percent of GRc,3.58 to 7.96 percent of GRb2,4.87 to 12.94 percent of GRb3 and 7.28 to 13.60 percent of GRd.
4. A method for preparing the active ginsenoside composition of any one of claims 1 to 3, comprising the steps of:
Dissolving total saponins of Ginseng radix, and loading into reversed phase C 18 In the silica gel chromatographic column, eluting with 43 volume percent of ethanol water solution, eluting with 50-55 volume percent of ethanol water solution when ginsenoside GRb1 is detected, collecting eluate until ginsenoside GRd is not detected by the eluate, and combining the collected eluate to obtain an active panaxadiol saponin composition, or detecting on-line fractions respectively collecting ginsenosides GRb1, GRc, GRb2, GRb3 and GRd and then mixing according to specific mass to obtain the active panaxadiol saponin composition;
or dissolving total saponins of Ginseng radix in reverse phase C 18 In the silica gel chromatographic column, 30% ethanol water solution is used for eluting firstly, 50-55% ethanol water solution is used for eluting when the ginsenoside GRg1 is detected, and the eluent is collected until the ginsenoside GRd is not detected, and the elution is stopped, and the collected eluent is combined to obtain the active holographic ginsenoside composition, or the ginsenoside GRg1 is detected and respectively collected on a combination line,The fractions of GRe, GRb1, GRc, GRb2, GRb3 and GRd are then mixed according to a specific mass to obtain an active holographic ginsenoside composition;
Or mixing total saponins of Ginseng radix as raw material to obtain active total ginsenoside composition.
5. The preparation method according to claim 4, wherein the solvent for dissolving the total saponins of the ginseng crude drug is 30% ethanol water solution by volume percent; the mass volume ratio of the total saponins of the ginseng crude drug to the 30% ethanol water solution is 1mg: 8-12 mL;
the method also comprises the following steps before loading: the reversed phase C is subjected to the process of adopting 30 percent ethanol water solution by volume percent 18 Performing balance treatment on the silica gel chromatographic column; the C is 18 The mass ratio of the usage amount of the silica gel to the total saponins of the raw medicinal materials is (7-10): 1.
6. the method of claim 4 or 5, wherein the ginseng crude drug or total saponins include, but are not limited to, the following combinations: the composition comprises a combination of American ginseng root or American ginseng root total saponin and American ginseng stem and leaf or American ginseng stem and leaf total saponin, a combination of ginseng root or American ginseng root total saponin and ginseng stem and leaf or ginseng stem and leaf total saponin, a combination of ginseng root or ginseng root total saponin, ginseng stem and leaf or ginseng stem and leaf total saponin, a combination of ginseng stem and leaf or ginseng stem and leaf total saponin, american ginseng stem and leaf or ginseng stem and leaf total saponin, a combination of American ginseng root or American ginseng root total saponin, american ginseng stem and leaf or American ginseng stem and leaf total saponin and notoginseng stem and leaf or ginseng stem and leaf total saponin;
The mass ratio of the raw materials or total saponins of each ginseng in each combination is determined according to the content of each ginsenoside contained in each raw material or total saponin which is actually used;
preferably, in the combination of the American ginseng root total saponins and the American ginseng stem and leaf total saponins, the mass ratio of the American ginseng root total saponins to the American ginseng stem and leaf total saponins is 1 (2-3);
the mass ratio of the ginseng root total saponins, the American ginseng root total saponins, the ginseng stem and leaf total saponins and the American ginseng stem and leaf total saponins in the combination of the ginseng root total saponins, the American ginseng root total saponins, the ginseng stem and leaf total saponins and the American ginseng stem and leaf total saponins is 1:1:1:3, a step of;
preferably, in the combination of the ginseng root total saponins, the ginseng stem and leaf total saponins and the notoginseng stem and leaf total saponins, the mass ratio of the ginseng root total saponins, the ginseng stem and leaf total saponins and the notoginseng stem and leaf total saponins is (1-2) 1 (1-2);
in the combination of the American ginseng root total saponins, the American ginseng stem and leaf total saponins and the pseudo-ginseng stem and leaf total saponins, the mass ratio of the American ginseng root total saponins to the American ginseng stem and leaf total saponins is 1:1 (1-2);
further preferably, the total saponins of the ginseng crude drug are extracted from the ginseng crude drug or are commercial products;
The total saponins of the ginseng crude drug extracted from the ginseng crude drug are prepared by mixing the ginseng crude drug and then using a conventional method, or the total saponins of the crude drugs are prepared by a conventional method respectively, and then the total saponins of the ginseng crude drug are obtained after mixing;
preferably, the extraction method comprises the steps of mixing the ginseng raw material, percolating and extracting for 3 times by using 70-90% ethanol water solution with the volume percentage content, and removing the solvent to obtain the ginseng raw material total saponins.
7. Use of the active ginsenoside composition according to any one of claims 1 to 3 or the active ginsenoside composition prepared by the preparation method according to any one of claims 4 to 6 in the preparation of a medicament for preventing and/or treating diseases or a health care product for exerting health care effects.
8. The use according to claim 7, characterized in that the active panaxadiol saponin composition and/or the active holographic ginsenoside composition is used for the preparation of a medicament for the prevention and/or treatment of a disease comprising at least one of the following: neurological disorders, autoimmune diseases, stress disorders and aging and related diseases;
preferably, the neurological condition comprises at least one of: mental disorders, sleep disorders, delayed and/or dysdevelopmental disorders, nerve injury and dysfunctional disorders, neurodegenerative disorders, addictive disorders and peripheral neuropathy;
Further preferably, the psychotic disorder comprises at least one of: schizophrenia, depression, bipolar disorder, anxiety and mania;
the sleep disorder includes at least one of: difficulty falling asleep, sleep maintenance disorder, early wake, reduced sleep quality, dreaminess, and reduced total sleep time; meanwhile, daytime dysfunction is accompanied, and daytime drowsiness, fatigue, inattention and hypomnesis occur, and tension, forcing and low emotion are accompanied;
the delayed and/or dysplastic condition comprises at least one of the following: hyperactivity, inattention, learning disorders, attention deficit hyperactivity disorder/hyperactivity disorder, autism, language disorders, sleep disorders, tourette syndrome/tourette syndrome, and tourette syndrome;
the nerve injury and dysfunctional disease includes at least one of: neuropsychiatric dysfunction and akinesia sequelae in epilepsy, acute and chronic phases of stroke;
the neurodegenerative disease comprises at least one of: parkinsonism, alzheimer's disease, vascular dementia, dementia of mixed type, secondary dementia, brain atrophy, chorea, multiple sclerosis and progressive freezing syndrome;
The parkinsonism comprises at least one of the following: multisystem degeneration-parkinsonism superposition, primary parkinsonism, juvenile parkinsonism, secondary parkinsonism caused by infection or ischemia, hereditary parkinsonism, and extrapyramidal systemic responses caused by drug therapy;
the multisystem degeneration-parkinsonism syndrome stack includes, but is not limited to, at least one of: multisystemic atrophy, progressive supranuclear palsy, corticobasal degeneration and frontotemporal lobar degeneration; the addictive disorders include at least one of the following: alcohol and drug addiction, juvenile networking and gaming addiction, and pathological gambling;
the peripheral neuropathy includes at least one of: neuralgia, facial neuritis, facial spasm, multiple peripheral neuropathy, guillain-Barre syndrome, and neuralgia and dyskinesia caused by viral infection;
preferably, the autoimmune disease comprises at least one of: lupus erythematosus, autoimmune glomerulonephritis, rheumatoid arthritis, dermatomyositis, scleroderma, allergic rhinitis, allergic asthma, urticaria, allergic conjunctivitis, demyelinating diseases, connective tissue diseases, neuromuscular diseases, digestive system diseases, endocrine diseases, and urinary system diseases;
Further preferred, the connective tissue disease comprises at least one of: systemic lupus erythematosus, rheumatoid arthritis, dermatomyositis and scleroderma;
the neuromuscular disease includes at least one of the following: multiple sclerosis, myasthenia gravis, and demyelinating diseases;
the digestive system disease includes at least one of: chronic non-specific ulcerative colitis, chronic active hepatitis and pernicious anaemia and atrophic gastritis;
the endocrinopathy includes at least one of the following: primary adrenocortical atrophy and chronic thyroiditis;
the urinary system diseases comprise autoimmune glomerulonephritis and/or pulmonary renal hemorrhagic syndrome;
preferably, the stress disorder comprises chronic stress-induced sub-health status and/or stress wound sequelae;
further preferred, the sub-health status includes insomnia, dreaminess and daytime dysfunction and its associated daytime sleepiness, fatigue weakness, inattention, hypomnesis, reduced work efficiency and creativity, or with anxiety, obsessive-compulsive, depressed mood;
further preferred, the stress wound sequelae comprises at least one of: acute stress disorders, maladaptation and post-traumatic stress disorders;
Preferably, the aging and its related diseases include at least one of the following: premature senility, senile hypomnesis, senile hypertension, senile sleep disorder, senile constipation and senile chronic inflammation.
9. The use according to claim 8, wherein the dosage form of the medicament comprises a liquid formulation and/or a solid formulation;
the liquid preparation comprises oral liquid and/or injection;
the solid formulation includes, but is not limited to, at least one of the following: tablets, capsules, granules, pills, enteric-coated preparations, controlled-release preparations and nano-preparations.
10. The use according to claim 8, characterized in that the use comprises the use of the active panaxadiol saponin composition or the active holographic ginsenoside composition as sole active ingredient and/or the active panaxadiol saponin composition or the active holographic ginsenoside composition in combination with the existing pharmaceutical preparation complex formulation exerts a synergistic attenuation effect on the existing pharmaceutical.
11. The use according to claim 10, wherein the existing medicament includes, but is not limited to, at least one of the following: levodopa and dopamine 2 receptor agonists for the treatment of parkinson's disease, dopamine receptor inhibitors for anti-schizophrenia and anxiety disorders, antiepileptic and neurological damage, sodium/calcium ion channel inhibitors for the treatment of neuropsychiatric behavioral disorders characterized by glutamate hyperexcitability or concomitant GABA-inhibitory deficiency, mycophenolate mofetil, antitumor chemotherapeutic or target agent for the treatment of autoimmune diseases and anti-organ transplant rejection;
Further preferably, the levodopa drug comprises: meadouba, xiding and Daling complex;
dopamine 2 receptor agonists include, but are not limited to: arvensis, ropinirole and cabergoline;
the inhibitors of the baamine receptor include, but are not limited to: haloperidol, olanzapine, clozapine, and risperidone;
the sodium and calcium ion channel inhibitors include, but are not limited to: lamotrigine or gabapentin;
when the active panaxadiol saponin composition and lamotrigine are used in combination or the compound medicine prepared by the active panaxadiol saponin composition and lamotrigine has synergistic attenuation function, the compound medicine can be used for treating epilepsy, bipolar disorder and acute cerebral ischemia injury, including but not limited to: cerebral ischemia of birth canal of newborn in acute stage of cerebral apoplexy;
when the active panaxadiol saponins and the gabapentin are combined or the compound medicine prepared by the active panaxadiol saponins and the gabapentin is used for exerting synergistic attenuation effect, the compound medicine is used for treating hypoevolutism or development disorder, alcohol and medicine addiction, teenager network and game addiction and pathological gambling;
such chemotherapeutic or target agents include, but are not limited to: paclitaxel and tyrosine kinase inhibitors.
12. The use according to claim 7, characterized in that the active panaxadiol saponin composition and/or the active holographic ginsenoside composition and/or the active total ginsenoside composition is used for preparing a health care product with health care functions, the health care functions comprising at least one of the following: delaying aging, improving health level and life quality of the elderly, improving sub-health state, and relieving nervous system side effects caused by drug treatment;
Preferably, the drug treatment causes side effects of the nervous system including side effects of the nervous system caused by neuropsychiatric drugs or anti-tumor drugs;
preferably, the neuropsychiatric drug comprises at least one of: dopamine receptor inhibitors and agonists, levodopa drugs, 5-hydroxytryptamine drugs, sodium-calcium channel inhibitors, glutamate receptor inhibitors, and other neurotransmitter receptor inhibitors;
the antitumor drug comprises at least one of the following: chemotherapeutic agents, molecular targeting agents, and immunotherapeutic agents;
preferably, the anti-aging comprises at least one of: inhibiting or preventing premature senility, improving senile hypomnesis, improving sleep, stimulating appetite, relieving senile chronic inflammation, improving senile mobility and prolonging life;
preferably, the sub-health state comprises at least one of the following states: insomnia, dreaminess, sleep disorders, tension, anxiety, hypomnesis, physical and mental fatigue, and reduced work efficiency.
13. The use according to claim 12, wherein the health product comprises a liquid health care product and/or a solid health care product;
The liquid health care product comprises oral liquid;
the solid health care product comprises at least one of: tablets, capsules, granules and pills.
14. A health product comprising the active total ginsenoside composition of claim 3 or prepared by combining at least one of the following ingredients: nutritional ingredients and active ingredients;
the nutritional ingredients include at least one of the following: proteins, polypeptides and glutathione precursor amino acids, NAD + Precursors and nucleic acids;
the active ingredients comprise at least one of the following: the medicine and food homology traditional Chinese medicine extract, coenzyme Q10, vitamins and energy metabolism intermediates.
15. The large health product of claim 14, wherein the large health product comprises an oral formulation;
the oral formulation comprises a solid formulation and/or a liquid formulation;
the solid formulations include, but are not limited to: capsules, normal tablets, dispersible tablets, enteric-coated tablets and granules.
CN202211636359.9A 2022-12-20 2022-12-20 Active ginsenoside composition and preparation method and application thereof Pending CN116019819A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211636359.9A CN116019819A (en) 2022-12-20 2022-12-20 Active ginsenoside composition and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211636359.9A CN116019819A (en) 2022-12-20 2022-12-20 Active ginsenoside composition and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN116019819A true CN116019819A (en) 2023-04-28

Family

ID=86090463

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211636359.9A Pending CN116019819A (en) 2022-12-20 2022-12-20 Active ginsenoside composition and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN116019819A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117323270A (en) * 2023-11-29 2024-01-02 江西康一参生物科技有限公司 Anti-inflammatory multifunctional ginseng composition and application thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101032535A (en) * 2006-12-22 2007-09-12 吉林人参研究院 Extracting purified ginsenoside from leaves of Panax quinquefolium and ginseng at the same time and the preparing method thereof
CN102727508A (en) * 2012-07-15 2012-10-17 浙江大学 Preparation of panaxadiol saponins component and pharmaceutical application for prevention and treatment of Parkinson disease
CN102743401A (en) * 2012-07-15 2012-10-24 浙江大学 Application of panaxadiol saponins fraction in preparing medicine for preventing epilepsia
KR101343819B1 (en) * 2013-04-05 2013-12-20 대동고려삼 주식회사 The composition for improving antistress, antihyperlipidemia and antiobesity control using red ginseng extract powder, vitamins and minerals, and the health functional food
US20140328951A1 (en) * 2011-10-07 2014-11-06 Gbiomix. Co., Ltd. Composition for Preventing and Treating Vision Deterioration and Age-Related Macular Degeneration through Retinal Repair Using Ginseng/Red Ginseng Extracts and Ginsenoside
CN104224817A (en) * 2014-08-29 2014-12-24 浙江大学 Application of panaxadiol saponin component in preparing antischizophrenic drug
US20220378862A1 (en) * 2021-05-26 2022-12-01 Daedong Korea Ginseng Co., Ltd Process for production of red ginseng hydrolysis concentrate having enriched specific component

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101032535A (en) * 2006-12-22 2007-09-12 吉林人参研究院 Extracting purified ginsenoside from leaves of Panax quinquefolium and ginseng at the same time and the preparing method thereof
US20140328951A1 (en) * 2011-10-07 2014-11-06 Gbiomix. Co., Ltd. Composition for Preventing and Treating Vision Deterioration and Age-Related Macular Degeneration through Retinal Repair Using Ginseng/Red Ginseng Extracts and Ginsenoside
CN102727508A (en) * 2012-07-15 2012-10-17 浙江大学 Preparation of panaxadiol saponins component and pharmaceutical application for prevention and treatment of Parkinson disease
CN102743401A (en) * 2012-07-15 2012-10-24 浙江大学 Application of panaxadiol saponins fraction in preparing medicine for preventing epilepsia
KR101343819B1 (en) * 2013-04-05 2013-12-20 대동고려삼 주식회사 The composition for improving antistress, antihyperlipidemia and antiobesity control using red ginseng extract powder, vitamins and minerals, and the health functional food
CN104224817A (en) * 2014-08-29 2014-12-24 浙江大学 Application of panaxadiol saponin component in preparing antischizophrenic drug
US20220378862A1 (en) * 2021-05-26 2022-12-01 Daedong Korea Ginseng Co., Ltd Process for production of red ginseng hydrolysis concentrate having enriched specific component

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117323270A (en) * 2023-11-29 2024-01-02 江西康一参生物科技有限公司 Anti-inflammatory multifunctional ginseng composition and application thereof
CN117323270B (en) * 2023-11-29 2024-03-19 江西康一参生物科技有限公司 Anti-inflammatory multifunctional ginseng composition and application thereof

Similar Documents

Publication Publication Date Title
Armentero et al. Past, present and future of A2A adenosine receptor antagonists in the therapy of Parkinson's disease
Sebastiao et al. Adenosine receptors and the central nervous system
Barriere et al. The restless legs syndrome
Liu et al. Icariin and icaritin ameliorated hippocampus neuroinflammation via inhibiting HMGB1-related pro-inflammatory signals in lipopolysaccharide-induced inflammation model in C57BL/6 J mice
Li et al. Antidepressant-like effects of the saponins extracted from Chaihu-jia-longgu-muli-tang in a rat unpredictable chronic mild stress model
WO2013079623A1 (en) Vicenin 2 and analogues thereof for use as an antispasmodic and/or prokinetic agent
Kim et al. Combined treatment with acupuncture reduces effective dose and alleviates adverse effect of L-dopa by normalizing Parkinson's disease-induced neurochemical imbalance
Takeda et al. Suppressive effect of Yokukansan on excessive release of glutamate and aspartate in the hippocampus of zinc-deficient rats
JP2017501975A (en) Application of ginsenoside Rg3 in the preparation of a medicament for preventing and / or treating dementia, and a medicament for treating dementia
Li et al. Ginsenoside Rb1 protects dopaminergic neurons from inflammatory injury induced by intranigral lipopolysaccharide injection
US9555055B2 (en) Use of albiflorin and metabolites thereof
Rastogi et al. Novel treatment modalities for painful diabetic neuropathy
CN116019819A (en) Active ginsenoside composition and preparation method and application thereof
Nam et al. KDS2010, a newly developed reversible MAO-B inhibitor, as an effective therapeutic candidate for Parkinson's disease
Li et al. Protective effect of Zhen-Wu-Tang (ZWT) through keeping DA stable and VMAT 2/DAT mRNA in balance in rats with striatal lesions induced by MPTP
Liu et al. α7 Nicotinic acetylcholine receptor: a key receptor in the cholinergic anti-inflammatory pathway exerting an antidepressant effect
Xie et al. Genistein-3′-sodium sulfonate ameliorates cerebral ischemia injuries by blocking neuroinflammation through the α7nAChR-JAK2/STAT3 signaling pathway in rats
Lu et al. Cortex Mori Radicis extract promotes neurite outgrowth in diabetic rats by activating PI3K/AKT signaling and inhibiting Ca2+ influx associated with the upregulation of transient receptor potential canonical channel 1 Corrigendum in/10.3892/mmr. 2020.11008
Liu et al. Novel synergistic mechanism of 11-keto-β-boswellic acid and Z-Guggulsterone on ischemic stroke revealed by single-cell transcriptomics
Liu et al. Optimal compatibility proportional screening of Trichosanthis Pericarpium-Trichosanthis Radix and its anti-Inflammatory components effect on experimental zebrafish and coughing mice
CN101837066A (en) Application of golden thread detoxication decoction as compound preparation in preparing IOD active medicaments
Chen et al. The globus pallidus as a target for neuropeptides and endocannabinoids participating in central activities
WO2019006734A1 (en) Use of (+)-2-borneol in preparation of drug for promoting upregulation of expression of sphingosine kinase-1 and/or bdnf
Zhao et al. PKM2-mediated neuronal hyperglycolysis enhances the risk of Parkinson's disease in diabetic rats
Zhu et al. Anticonvulsant effects of dingxian pill in pentylenetetrazol-kindled rats

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination