CN117085138A - Composition for improving immunity and resisting pathogenic infection and application thereof - Google Patents

Composition for improving immunity and resisting pathogenic infection and application thereof Download PDF

Info

Publication number
CN117085138A
CN117085138A CN202311141947.XA CN202311141947A CN117085138A CN 117085138 A CN117085138 A CN 117085138A CN 202311141947 A CN202311141947 A CN 202311141947A CN 117085138 A CN117085138 A CN 117085138A
Authority
CN
China
Prior art keywords
alox15
composition
acid
virus
inhibitor
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
CN202311141947.XA
Other languages
Chinese (zh)
Inventor
李怡芳
何蓉蓉
栗原博
欧阳淑桦
张琼谊
翁竞玉
陈新星
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.)
Jinan University
Original Assignee
Jinan University
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 Jinan University filed Critical Jinan University
Priority to CN202311141947.XA priority Critical patent/CN117085138A/en
Publication of CN117085138A publication Critical patent/CN117085138A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7032Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a polyol, i.e. compounds having two or more free or esterified hydroxy groups, including the hydroxy group involved in the glycosidic linkage, e.g. monoglucosyldiacylglycerides, lactobionic acid, gangliosides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The application belongs to the field of medicines, and in particular relates to a composition for improving natural immunity and playing an anti-infection role and application thereof. Specifically, the application selects lipoxygenase ALOX15 enzyme activity inhibitor including baicalein, compound with ALOX15 transcription promoting activity including cynarone, and compound with ALOX15 protein degradation inhibiting effect including forsythoside A/E/I, and carries out multi-strategy combined scientific formula compatibility. The experimental study shows that the composition containing the ALOX15 enzyme activity inhibitor, the ALOX15 transcriptional activity promoter and the ALOX15 protein degradation inhibitor can well improve the natural immunity of organisms, increase the synthesis and secretion of IFN-beta and play a role in resisting pathogenic infection. The composition can also inhibit the production of pathogenic-induced inflammatory factors, has a therapeutic effect on pathogenic-induced secondary inflammatory symptoms, and has good clinical application prospect.

Description

Composition for improving immunity and resisting pathogenic infection and application thereof
Technical Field
The application belongs to the field of medicines, and particularly relates to a composition for improving immunity and resisting pathogenic infection and application thereof.
Background
Infectious diseases are caused by a number of pathogens such as bacteria, fungi, rickettsiae, viruses, parasites, and the like, causing inflammatory storms, resulting in tracheitis, bronchitis, pneumonia, arthritis, otitis media, and even other gastrointestinal diseases and neurological symptoms. The natural immune system is used as a first line of defense of the body against infection by external pathogens, and is activated first after the body is infected, and then recognizes and clears the corresponding pathogens. The response of type I interferon IFN, particularly IFN- β signaling, to pathogens during innate immune activation in the body is an important anti-infective mechanism for the body. Therefore, increasing IFN levels in the body and improving immunity in the body are important strategies against pathogenic infection.
The inventors have found that 15-lipoxygenase (ALOX 15) has strong anti-infective activity, and that ALOX15 exerts anti-infective effect in the body independent of its enzymatic activity. Further, the inventors have found that ALOX15 levels are positively correlated with the levels of the innate anti-infective agent IFN- β in the body, i.e., increasing expression of ALOX15 promotes synthesis and secretion of IFN- β. Therefore, ALOX15 can be used as a novel anti-infection target to develop novel anti-pathogenic infection medicines, health products or functional foods.
At present, anti-infection Chinese and Western medicine treatment medicines and methods aiming at pathogens including various viruses, bacteria, fungi, parasites and the like are numerous, but the existing medicines have the defects of low efficiency, obvious adverse reaction, even lack of effective medicines and the like whether aiming at novel viruses, influenza viruses, host long-term symbiotic viruses, bacteria, fungi, parasites and the like. Thus, there is an urgent need to develop new therapeutic methods and medicaments. The traditional Chinese medicine composition or the natural product composition reported in the prior published patent and related literature mostly has the defects of undefined treatment target, complex composition, complex source, high cost and the like, and greatly limits the clinical transformation of the traditional Chinese medicine composition or the natural product composition.
Therefore, the application is based on the previous research results of the inventor, takes ALOX15 as a drug development target, selects a compound which has the functions of promoting the transcriptional activity of the ALOX15, inhibiting the degradation of ALOX15 protein and inhibiting the enzymatic activity of the ALOX15, carries out scientific compatibility, increases the protein level of the ALOX15 at the organism or focus part and inhibits the lipid peroxidation level of the organism or focus part at multiple times, and has definite development target, good curative effect, small toxic and side effect and low cost. The composition has effects of enhancing immunity, resisting pathogenic infection, and inhibiting inflammation.
Disclosure of Invention
In order to solve the technical problems, the application aims to provide an active ingredient composition for increasing the ALOX15 protein content and inhibiting the enzyme activity of the active ingredient composition and a new application of the active ingredient composition in the aspects of enhancing the immunity of organisms and resisting pathogenic infection.
Specifically, the application is realized through the following technical schemes:
the present application provides the use of a composition comprising an inhibitor of ALOX15 enzyme activity, an promoter of ALOX15 transcriptional activity and/or an inhibitor of ALOX15 protein degradation, and/or other inhibitors of protein degradation, in the manufacture of a medicament for enhancing immunity and against pathogenic infection.
Alternatively, in the above use, the ALOX15 protein degradation inhibitor is selected from an ALOX15 protein ubiquitination degradation inhibitor, and/or an ALOX15 protein autophagy degradation inhibitor, and/or other protein degradation inhibitors.
Alternatively, in the above use, the composition further comprises one or more additional active ingredients selected from the group consisting of: neochlorogenic acid (5-O-caffeoylquinic acid), semen Strychni glycoside (secoogenin), forsythin, caffeic acid, chlorogenic acid, isochlorogenic acid, caffeine, ursodeoxycholic acid, baicalin, quercetin, rosmarinic acid, or derivatives thereof, or Chinese medicinal materials and/or extracts containing the same, or any combination thereof.
Alternatively, in the above use, the composition further comprises one or more inflammation-inhibiting active ingredients selected from the group consisting of: ursodeoxycholic acid, atractylone, peony ketone, perillaldehyde, or derivatives thereof, chinese medicinal materials and/or extracts containing the above compounds, or any combination thereof.
Preferably, in the above use, the ALOX15 enzyme activity inhibitor is selected from one or more of the following: baicalein, rosmarinic acid, quercetin, chlorogenic acid, isochlorogenic acid, caffeic acid, caffeine, PD146176, daidzein, phellodendrine, ferulic acid, hesperetin, hesperidin, ML351, or derivatives thereof, or Chinese medicinal materials and/or extracts containing the above compounds, or any combination thereof.
Preferably, in the above use, the ALOX15 transcriptional activity promoter is selected from one or more of the following: the preparation comprises Songarine (songorine), neochlorogenic acid, and strychnine oxide, or derivatives thereof, and Chinese medicinal materials and/or extracts containing the above compounds, or any combination thereof.
Preferably, in the above use, the ALOX15 protein degradation inhibitor is selected from one or more of the following: forsythoside A, forsythoside E, forsythoside I, or derivatives thereof, chinese medicinal materials and/or extracts containing the same, or any combination thereof.
Preferably, as an alternative, in the above use, the composition mainly comprises the aconitine, baicalein and forsythoside E.
Preferably, as an alternative, in the above use, the composition consists of the aconitine, baicalein and forsythoside E.
Alternatively, in the above use, the active ingredient is artificially synthesized and/or extracted from a plant, and/or a natural product modified product.
Alternatively, in the above use, the pathogen is selected from one or more of the following pathogenic microorganisms: influenza virus, common coronavirus, novel coronavirus (covd-19), SARS virus, MERS virus, hepatitis c virus, encephalitis B virus, rhinovirus, polio virus, coxsackie virus, dengue virus, rotavirus, ebola virus, aids virus, marburg virus, mycobacterium tuberculosis, legionella pneumophila, pathogenic escherichia coli, salmonella typhimurium, streptomyces fradiae, group B streptococcus, staphylococcus aureus, pneumococcus, candida albicans, aspergillus fumigatus, chlamydia trachomatis, chlamydia pneumoniae, or any combination thereof.
Preferably, in the above use, the pathogen is influenza virus, common coronavirus, novel coronavirus, mycobacterium tuberculosis, chlamydia trachomatis or any combination thereof.
Alternatively, in the above use, the composition may further prevent or treat a disease associated with the pathogenic infection, which is an inflammatory disease caused by the pathogenic infection, selected from one or more of the following: pneumonia and its complications, tracheitis, bronchitis, laryngitis, encephalitis, pharyngitis, nephritis, hepatitis, enteritis, gastritis, arthritis, conjunctivitis, otitis media, or any combination thereof.
Alternatively, in the above use, the composition maintains the protein level of the antipathogenic infectious protein ALOX15 and inhibits its enzymatic activity, promotes synthesis and secretion of the innate immune factor IFN- β, enhances innate immunity, inhibits pathogen invasion into the host, inhibits host inflammation.
Alternatively, in the above use, the composition inhibits the expression/secretion of at least one of inflammatory factors TNF- α, IL-6, IL-1 β, IFN- γ, and/or reduces the number of inflammatory infiltrating cells.
Alternatively, in the above application, the composition is added with conventional adjuvants, and made into clinically acceptable medicines, or commercially acceptable health products or functional foods according to conventional process. The dosage form of the medicine is selected from one of liquid preparation, solid preparation or semisolid preparation.
Preferably, the medicament is in the form of gel, cream, tablet, capsule, powder, mixture, pill, granule, oral liquid, syrup, extract, suppository, aerosol, patch, ointment, injection, spray, liniment, tincture, wet compress, paste or lotion.
Compared with the prior art, the application has the following beneficial effects:
(1) The application takes ALOX15 as a drug development target point, firstly selects a compound which has the functions of promoting the transcriptional activity of the ALOX15, inhibiting the degradation of ALOX15 protein and inhibiting the enzymatic activity of the ALOX15, carries out scientific compatibility, increases the protein level of the ALOX15 of an organism or a focus part and inhibits the lipid peroxidation level of the organism or the focus part simultaneously, and successfully develops a drug composition which has definite target point, good curative effect, small toxic and side effect, low cost, and has the functions of enhancing immunity, resisting pathogenic infection, inhibiting inflammation and the like.
(2) Pharmacological experiments prove that various active ingredients in the pharmaceutical composition have remarkable synergistic effects in the aspects of enhancing immunity, resisting pathogenic infection, inhibiting inflammation and the like, have no toxic or side effect, and have good clinical application prospect.
(3) The various natural product monomers used in the pharmaceutical composition have wide sources and can be produced by a mature method, so that the pharmaceutical composition is convenient for industrialized mass production. In addition, the pharmaceutical composition is prepared into a conventional preparation by adding a pharmaceutically acceptable carrier, so that the curative effect of the medicine is stable and the use is convenient.
Drawings
The accompanying drawings are included to provide a further understanding of the application and are incorporated in and constitute a part of this specification, illustrate the application and together with the embodiments of the application, serve to explain the application.
In the drawings:
fig. 1: the composition of the application increases the survival rate of mice infected with influenza virus.
Fig. 2: the composition of the application reduces the lung index of influenza virus infected mice.
Fig. 3: the composition of the application reverses lung injury caused by influenza virus infection.
Fig. 4: the composition of the application inhibits the secretion of inflammatory factors caused by influenza viruses.
Fig. 5: the compositions of the application promote IFN- β secretion in mice.
Fig. 6: the composition of the application promotes IFN-beta secretion of mycobacterium tuberculosis and chlamydia trachomatis infected cells.
Fig. 7: the compositions of the application increase ALOX15 protein levels.
Detailed Description
The application will be further illustrated with reference to specific examples. It should be understood that the detailed description and specific examples are intended for purposes of illustration only and are not intended to limit the scope of the application.
The specific techniques or conditions are not identified in the examples and are described in the literature in this field or are carried out in accordance with the product specifications. The reagents or equipment used were conventional products available for purchase through regular channels, with no manufacturer noted.
The experimental methods in the following examples are conventional methods unless otherwise specified. The test materials used in the examples described below, unless otherwise specified, are all commercially available products.
Example 1: in vitro anti-influenza virus Activity of the compositions of the present application
The following drugs were administered to lung cancer human alveolar basal epithelial (a 549) cells: the effect of the compound of the present application on the compound of the present application was composed of the following ingredients, namely, the compound of the present application was composed of the compound of the present application, either of the compound of the present application (10. Mu.g/mL) or of the compound of the present application, either of the compound of the present application (5. Mu.g/mL) or of the compound of the present application (5. Mu.g/mL), either of the compound of the present application (5. Mu.g/mL) or of the compound of the present application (2:1) or of the compound of the present application) (ursolic acid+isochlorogenic acid, 1:1) or of the compound of the present application (ursolic acid+isochlorogenic acid, 1:1). After 12 hours, the influenza virus H1N1 was adsorbed at 10 times TCID50 for 2 hours, and after 2 hours, the unadsorbed virus was washed off and then cultured continuously for 24 hours. 24 hours after virus infection, each set of genes was collected for RT-qPCR detection of NP levels. The synergy index is determined by adopting a gold positive average q value method, and q=e is obtained by the following formula A+B/ (E A +E B -E A ×E B ). In E A 、E B The inhibition rates of the aconitine group and the forsythoside E group, the inhibition rates of the A composition group and the baicalein group, or the inhibition rates of the B composition group and the C composition group are respectively. q<1 illustrates antagonism after the two drugs are combined; q>1 illustrates the synergistic effect of the two drugs combined, and q=1 illustrates the additive effect of the two drugs combined.
The experimental results are shown in table 1, the composition a can obviously inhibit the replication of influenza viruses in vitro, and the inhibition effects of the aconitine and the forsythoside E are lower than those of the composition a at the same dosage. In addition, the effect of the composition B on inhibiting virus replication is better than that of the composition A and baicalein. Composition D was optimal with a viral replication inhibition rate as high as 95.51%. The above results demonstrate that the compositions of the present application are the result of the synergistic interaction of the components in terms of their antiviral effect.
Table 1: in vitro anti-influenza virus Activity of the compositions of the present application
Note that: data are expressed in mean±sd, data statistics were performed using one-way ANOVA analysis of SPSS25.0, compared to normal control, * P<0.05, ** P<0.01, *** P<0.001。
example 2: protection of mice infected with influenza virus by the composition of the present application
1. The composition of the application increases the survival rate of mice infected by influenza virus
Male BALB/c mice of 6-8 weeks were selected and divided into 4 groups, which were respectively a normal control group, a model control group, and a group of (weight ratio of 2:1:1, group A), a group of (weight ratio of) cynarone, forsythoside E+baicalin+baicalin+ursodeoxycholic acid+isochlorogenic acid (weight ratio of 2:1:0.5:0.5, group B), and 10 mice each. Group A (composition A) and group B (composition B) mice were each given the corresponding composition solutions (10 mg/kg) by gavage, and the remaining groups of mice were given equal amounts of distilled water by gavage for 10 consecutive days. After 10 days, each of the other groups except the normal control group was infected with influenza A/FM/1/47 (H1N 1) strain with a 2-fold LD50 viral load by nasal drop. The mice in each group were continuously observed for 21 days and the survival was recorded.
As shown in fig. 1, the compositions of group a and group B significantly increased the survival rate of mice infected with the virus, and the composition of group B had better effect than the single virus infected group.
2. The composition of the application reduces the lung index of mice infected with influenza virus
Male BALB/c mice of 6-8 weeks were selected and divided into 4 groups, which were respectively a normal control group, a model control group, and a group of (weight ratio of 2:1:1, group A), a group of (weight ratio of) cynarone, forsythoside E+baicalin+baicalin+ursodeoxycholic acid+isochlorogenic acid (weight ratio of 2:1:0.5:0.5, group B), and 10 mice each. The group A and group B mice were respectively given the corresponding composition solutions (10 mg/kg) by gavage, and the remaining groups were given the same amount of distilled water by gavage for 10 days. After 10 days, each of the other groups except the normal control group was infected with influenza A/FM/1/47 (H1N 1) strain with a 2-fold LD50 viral load by nasal drop. Mice were weighed and recorded on day 5 of infection with H1N1 by nasal drop. Immediately thereafter, each group of mice was anesthetized with isoflurane, and lung tissue was weighed to calculate lung index. The lung index calculation formula is lung index=wet lung weight (g)/body mass (g) ×100%.
Data were plotted using Graphpad Prism software (version 8) and expressed as mean ± SD. Data statistics were analyzed for one-way variance using SPSS software (version 25), and the statistics met the criteria of a normal distribution of variance similarity.
The experimental results are shown in fig. 2, and the lung index of the model control group is obviously increased compared with that of the normal control group. After the composition is administered for protection, the lung index of the mice is obviously reduced.
3. The composition of the application reverses lung injury caused by influenza virus infection
Male BALB/c mice of 6-8 weeks were selected and divided into 4 groups, which were respectively a normal control group, a model control group, and a group of (weight ratio of 2:1:1, group A), a group of (weight ratio of) cynarone, forsythoside E+baicalin+baicalin+ursodeoxycholic acid+isochlorogenic acid (weight ratio of 2:1:0.5:0.5, group B), and 10 mice each. The group A and group B mice were respectively given the corresponding composition solutions (10 mg/kg) by gavage, and the remaining groups were given the same amount of distilled water by gavage for 10 days. After 10 days, each of the other groups except the normal control group was infected with influenza A/FM/1/47 (H1N 1) strain with a 2-fold LD50 viral load by nasal drop. On day 5 of H1N1 infection by nasal drops, mice of each group were anesthetized with isoflurane, and lung tissue was perfused and fixed in paraformaldehyde. Paraffin embedding, sectioning, HE staining was then performed to observe pathological changes in lung tissue.
The experimental results are shown in fig. 3, and compared with the normal control group, the model control group mice have lung injury including tissue necrosis and extensive inflammatory cell infiltration, and the injury condition of the lung tissue is obviously improved after the composition is protected.
4. The composition of the application inhibits the secretion of inflammatory factors caused by influenza viruses
Male BALB/c mice of 6-8 weeks were selected and divided into 4 groups, which were respectively a normal control group, a model control group, and a group of (weight ratio of 2:1:1, group A), a group of (weight ratio of) cynarone, forsythoside E+baicalin+baicalin+ursodeoxycholic acid+isochlorogenic acid (weight ratio of 2:1:0.5:0.5, group B), and 10 mice each. The group A and group B mice were respectively given the corresponding composition solutions (10 mg/kg) by gavage, and the remaining groups were given the same amount of distilled water by gavage for 10 days. After 10 days, each of the other groups except the normal control group was infected with influenza A/FM/1/47 (H1N 1) strain with a 2-fold LD50 viral load by nasal drop. On day 5 of H1N1 infection by nasal drop, each group of mice was anesthetized with isoflurane and mouse serum was collected for ELISA detection of IL-1. Beta. Levels.
Data were plotted using Graphpad Prism software (version 8) and expressed as mean ± SD. Data statistics were analyzed for one-way variance using SPSS software (version 25), and the statistics met the criteria of a normal distribution of variance similarity.
The results of the experiment are shown in FIG. 4, and the composition of the present application can significantly inhibit the IL-beta level.
Example 3: in vitro antiviral Activity of the compositions of the application against coronaviruses
Mixing the cynarophylline, the forsythoside E, the baicalein, the ursodeoxycholic acid and the isochlorogenic acid according to the weight ratio of 2:1:1:0.5:0.5, weighing and dissolving the mixture by using DMSO to perform an in-vitro anti-coronavirus drug effect experiment of the composition. Vero cells (african green monkey kidney cell line) were plated uniformly into 96-well plates and after 12 hours, 100 μl of 2-fold gradient diluted composition was added to each well. The blank and normal cell groups were added to an equal volume of culture medium and incubated under an incubator for 48 hours. Subsequently, 20. Mu.L of MTT solution at a concentration of 5mg/mL was added to each well and incubation was continued for 4 hours. The supernatant was discarded, 100. Mu.L of DMSO was added to each well, and the mixture was shaken at low speed for 5 minutes to charge the crystalsAnd (5) dissolving the components. The absorbance at 570nm was measured with an ultraviolet spectrophotometer, and the drug median toxicity concentration (TC 50 )。
Vero cells (African green monkey kidney cell line) were plated uniformly in 96-well plates at 100-fold TCID after 12 hours 50 The human coronavirus hCoV-229E viral load of (C) infected cells and adsorbed for 2 hours, after which unadsorbed virus was washed off. Then, 2-fold gradient dilutions of the composition extract were added, and 4 duplicate wells were set for each concentration, with the maximum non-toxic concentration being the initial concentration of drug, and the culture was continued in an incubator for 48 hours. Cytopathy (CPE) was recorded daily. OD value determination was performed and drug-calibrated effective concentration (IC) was calculated using MTT staining 50 ) The calculation formula for calculating the selection index SI, SI is as follows: si=tc 50 /IC 50 [ reference to the methods of pharmacological experiments, main code Xu Shuyun ]]Therapeutic index SI>1 denotes active.
The experimental results are shown in Table 2, and the composition has remarkable inhibition effect on human coronavirus hCoV-229E.
Table 2: inhibition of human coronavirus hCoV-229E by the compositions of the present application
Example 4: the composition of the application improves the immunity of mice infected by viruses
Male BALB/c mice of 6-8 weeks were selected and divided into 4 groups, which were respectively a normal control group, a model control group, and a group of (weight ratio of 2:1:1, group A), a group of (weight ratio of) cynarone, forsythoside E+baicalin+baicalin+ursodeoxycholic acid+isochlorogenic acid (weight ratio of 2:1:0.5:0.5, group B), and 10 mice each. The group A and group B mice were respectively given the corresponding composition solutions (10 mg/kg) by gavage, and the remaining groups were given the same amount of distilled water by gavage for 10 days. After 10 days, each of the other groups except the normal control group was infected with influenza A/FM/1/47 (H1N 1) strain with a 2-fold LD50 viral load by nasal drop. On day 5 of H1N1 infection by nasal drops, each group of mice was anesthetized with isoflurane and mouse serum was collected for ELISA detection of IFN- β levels.
Data were plotted using Graphpad Prism software (version 8) and expressed as mean ± SD. Data statistics were analyzed for one-way variance using SPSS software (version 25), and the statistics met the criteria of a normal distribution of variance similarity.
The results of the experiment are shown in FIG. 5, in which the composition of the present application promotes IFN- β secretion.
Example 5: the composition of the application promotes IFN-beta secretion of mycobacterium tuberculosis and chlamydia trachomatis infected cells
(1) Extraction of mouse bone marrow monocyte-derived macrophages BMDM: collecting C57BL/6 mice of 8 weeks old, removing neck, killing, placing into a tray containing 75% ethanol, soaking for 5 min, transferring into a super clean bench, cutting skin under aseptic condition, taking out femur and tibia of hind leg, and stripping muscle tissue. The femur and tibia were placed in a petri dish with sterile PBS. After the two ends of femur and tibia are sheared off, a proper amount of PBS is sucked by a 5mL syringe, 1mL of thin needle is replaced, the bone is pinched by forceps for the left hand, the needle is controlled by the right hand to be inserted into the marrow cavity of the femur and tibia and washed, a 50mL centrifuge tube for placing a cell filter screen is adopted to filter and collect PBS containing cells, and the PBS is washed until the bone cavity is colorless. The filtrate was dispensed into 15mL centrifuge tubes and centrifuged at 1200 rpm for 10 minutes. The supernatant was discarded, resuspended in 1mL medium and pooled into a tube, and 10. Mu.L of the cell suspension was counted in a cell counting plate. Counting the number of cells to 10 5 Each mL was plated and cultured in DMEM complete medium containing 50ng/mL M-CSF.
(2) BMDM cells infected with mycobacterium tuberculosis: mycobacterium tuberculosis H37Rv was infected with the BMDM cells obtained as described above at MOI 3 (multiplicity of infection ) 3 for 4 hours, while adding composition A (20. Mu.g/mLl) and composition B (20. Mu.g/mLl) to the cell culture medium, respectively, wherein composition A and composition B were present in the medium until IFN- β content was detected. The medium was then discarded and the extracellular bacteria were washed off with PBS and the medium containing the cells in gentamicin at 100. Mu.g/mL was replaced. Finally, at 20 hours post-infection (20 hpi), the culture supernatant was collected and the IFN- β levels in the supernatant were detected using ELISA detection kits.
(3) HeLa cells infected with chlamydia trachomatis (Ct): heLa cells were inoculated into 10cm dishes and cultured overnight in a cell incubator with DMEM medium containing 10% fetal bovine serum. The next day, when the cell density was about 80%, the medium was discarded, ct suspension was added, and infection was performed for 24 hours, while composition A (20. Mu.g/mL) and composition B (20. Mu.g/mL) were added to the cell medium, respectively, wherein composition A and composition B were present in the medium until IFN- β content was detected. After 24 hours, the medium was discarded, and the culture was continued for 24 hours by adding a growth solution (containing 10% fetal bovine serum and 2mg/L cycloheximide). Finally, the cell supernatant was collected and the IFN- β level in the supernatant was detected using ELISA detection kit.
Data were plotted using Graphpad Prism software (version 8) and expressed as mean ± SD. Data statistics were analyzed for one-way variance using SPSS software (version 25), and the statistics met the criteria of a normal distribution of variance similarity.
The results of the experiment are shown in FIG. 6, in which the composition of the present application promotes IFN- β secretion in Mycobacterium tuberculosis, chlamydia trachomatis-infected cells.
Example 6: the compositions of the application increase ALOX15 protein levels
The inventor has found in the early stage that ALOX15 can stabilize mitochondrial antiviral protein MAVS, inhibit ubiquitination degradation of the mitochondrial antiviral protein MAVS and promote the generation of antiviral molecule IFN-beta. In the present application, it was found that the composition of the present application (cynaro songaricum aconitine + forsythoside E + baicalin, weight ratio 2:1:1) increased the effect of ALOX15 protein levels.
The following drugs were administered to lung cancer human alveolar basal epithelial (a 549) cells: composition (aconitine, forsythoside E, baicalin, weight ratio of 2:1:1) (40 μg/mL), protein synthesis inhibitor cycloheximide (CHX, 100 μM), CHX+ composition for 12 hr. A group of normal controls was also set up and given the same volume of DMSO for the same time. After 12 hours, the cells were collected to extract proteins and ALOX15 protein levels were detected using western blot.
The results of the experiment are shown in FIG. 7, in which the composition of the present application significantly increases ALOX15 protein levels, indicating that the composition of the present application may exert an immune enhancing and antiviral effect through ALOX 15.
In conclusion, the composition of the application containing the ALOX15 enzyme activity inhibitor, the ALOX15 transcriptional activity promoter and the ALOX15 protein degradation inhibitor can well improve antiviral immunity, inhibit viral replication, increase synthesis and secretion of IFN-beta and play an antiviral role.
It will be apparent to those skilled in the art that various modifications and variations can be made to the present application without departing from the spirit or scope of the application. Thus, it is intended that the present application also include such modifications and alterations insofar as they come within the scope of the appended claims or the equivalents thereof.

Claims (9)

1. Use of a composition comprising an inhibitor of ALOX15 enzyme activity, an promoter of ALOX15 transcriptional activity and an inhibitor of ALOX15 protein degradation in the manufacture of a medicament for enhancing immunity and combating pathogenic infection.
2. Use according to claim 1, characterized in that: the ALOX15 protein degradation inhibitor is selected from ALOX15 protein ubiquitination degradation inhibitor and/or ALOX15 protein autophagy degradation inhibitor and/or other protein degradation inhibitor.
3. Use according to claim 1, characterized in that: the ALOX15 enzyme activity inhibitor is selected from one or more of the following: baicalein, rosmarinic acid, quercetin, chlorogenic acid, isochlorogenic acid, caffeic acid, caffeine, PD146176, daidzein, phellodendrine, ferulic acid, hesperetin, hesperidin, ML351 or Chinese medicinal materials and/or extracts containing the above compounds, or any other compound with ALOX15 enzyme activity inhibiting effect.
4. Use according to claim 1, characterized in that: the ALOX15 transcriptional activity promoter is selected from one or more of the following: the alkaloid (songorine), neochlorogenic acid, strychnine, or Chinese medicinal materials and/or extract containing the above compounds, or any other compound with ALOX15 transcriptional activity promoting effect.
5. Use according to claim 1, characterized in that: the ALOX15 protein degradation inhibitor is selected from one or more of the following: forsythoside A, forsythoside E, forsythoside I, or Chinese medicinal materials and/or extracts containing the above compounds, or any other compound with ALOX15 protein degradation inhibiting effect.
6. Use according to claim 1, characterized in that: the composition comprises one or more active ingredients selected from the group consisting of: neochlorogenic acid (5-O-caffeoylquinic acid), sepoxygenanin, forsythin, caffeic acid, chlorogenic acid, isochlorogenic acid, caffeine, ursodeoxycholic acid, caffeic acid, baicalin, quercetin, rosmarinic acid, or other compounds of any one of claims 3 to 5.
7. Use according to any one of claims 1-6, characterized in that: the active ingredient is an artificially synthesized and/or extracted from plants and/or natural product modified product.
8. Use according to claim 1, characterized in that: the pathogen is selected from one or more of the following pathogenic microorganisms: influenza virus, common coronavirus, novel coronavirus (covd-19), SARS virus, MERS virus, hepatitis c virus, encephalitis B virus, rhinovirus, polio virus, coxsackie virus, dengue virus, rotavirus, ebola virus, aids virus, marburg virus, mycobacterium tuberculosis, legionella pneumophila, pathogenic escherichia coli, salmonella typhimurium, streptomyces fradiae, group B streptococcus, staphylococcus aureus, pneumococcus, candida albicans, aspergillus fumigatus, chlamydia trachomatis, chlamydia pneumoniae, or any combination thereof.
9. Use according to any one of claims 1-8, characterized in that: the composition is added with conventional auxiliary materials and is prepared into clinically acceptable pharmaceutical dosage forms according to a conventional process.
CN202311141947.XA 2023-09-06 2023-09-06 Composition for improving immunity and resisting pathogenic infection and application thereof Pending CN117085138A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311141947.XA CN117085138A (en) 2023-09-06 2023-09-06 Composition for improving immunity and resisting pathogenic infection and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311141947.XA CN117085138A (en) 2023-09-06 2023-09-06 Composition for improving immunity and resisting pathogenic infection and application thereof

Publications (1)

Publication Number Publication Date
CN117085138A true CN117085138A (en) 2023-11-21

Family

ID=88783369

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311141947.XA Pending CN117085138A (en) 2023-09-06 2023-09-06 Composition for improving immunity and resisting pathogenic infection and application thereof

Country Status (1)

Country Link
CN (1) CN117085138A (en)

Similar Documents

Publication Publication Date Title
AU2011315708B2 (en) Formulations for the treatment of disorders of the upper respiratory tract
KR101317318B1 (en) A composition comprising the extract of Galla Rhois or the compounds isolated therefrom showing inhibiting activity of novel influenza, avian influenza, or SARS syndrome
CN113491703B (en) Application of phenylethanoid glycosides compounds and composition thereof in preparation of medicines for preventing and treating new coronavirus infection
CN106668832B (en) Application of polypeptide in preparation of medicine for treating enterovirus infection
CN111759851B (en) Application of tannic acid in preparing anti-coronavirus medicine
KR20210014601A (en) Composition for preventing or treating coronavirus infection
JP2011522038A (en) Triterpenoid compounds useful as virus inhibitors
CN111588744A (en) Antiviral composition and oral liquid as well as preparation method and application thereof
JP2023522644A (en) Therapeutic use of pleuromutilin
CN117085138A (en) Composition for improving immunity and resisting pathogenic infection and application thereof
CN114748466B (en) Application of Napyradiomycin compounds in preparation of porcine pseudorabies virus inhibitor
CN112843073A (en) Application of Reddesivir (Remdesivir) in preparation of anti-bovine parainfluenza virus type 3 medicine
CN110974819B (en) Composition for upper respiratory tract infection and application thereof
CN114377027A (en) Application of pulsatilla saponin B4 in preparation of medicine for treating or preventing SARS-CoV-2
CN112546202A (en) Complexing agent with HPV virus inhibiting function and preparation method thereof
CN113304158B (en) Application of sulfamethoxypyrazine in preparation of product for preventing and/or treating bovine parainfluenza virus
CN113318115B (en) Application of metacycline in preparation of medicine for preventing and treating bovine parainfluenza virus type 3 virus infection
CN114917285B (en) Application of polysaccharide compound in medicine for preventing and treating respiratory tract virus infection
KR101665015B1 (en) Composition for the prevention and treatment of antiviral comprising extracts of crude drug complex
KR101665016B1 (en) Composition for the prevention and treatment of antiviral comprising extracts of crude drug complex
CN117165607A (en) Gene therapy method for enhancing immunity and preventing and treating infectious diseases and complications thereof
CN112691094B (en) Novel compound for preventing and treating virus and application thereof
CN112353809B (en) Pharmaceutical application of astragaloside IV compound
CN113797193B (en) Application of trimethoxy quercetin in preparation of anti-coronavirus medicines
CN110179785B (en) Application of widmanone in preparation of medicine for treating or preventing hand-foot-and-mouth disease

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