US20100022631A1 - Treatment of hepatic encephalopathy and liver cirrhosis - Google Patents

Treatment of hepatic encephalopathy and liver cirrhosis Download PDF

Info

Publication number
US20100022631A1
US20100022631A1 US12/429,832 US42983209A US2010022631A1 US 20100022631 A1 US20100022631 A1 US 20100022631A1 US 42983209 A US42983209 A US 42983209A US 2010022631 A1 US2010022631 A1 US 2010022631A1
Authority
US
United States
Prior art keywords
treatment
liver
taa
capsaicin
hepatic encephalopathy
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.)
Abandoned
Application number
US12/429,832
Inventor
Elliot Berry
Yosefa Avraham
Raphael Mechoulam
Yaron Ilan
Yossi Dagon
Iddo Magen
Nicholas Grigoriadis
Theofilos Poutachidis
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.)
Aristotle University of Thessaloniki
Hadasit Medical Research Services and Development Co
Yissum Research Development Co of Hebrew University of Jerusalem
Original Assignee
Aristotle University of Thessaloniki
Hadasit Medical Research Services and Development Co
Yissum Research Development Co of Hebrew University of Jerusalem
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 Aristotle University of Thessaloniki, Hadasit Medical Research Services and Development Co, Yissum Research Development Co of Hebrew University of Jerusalem filed Critical Aristotle University of Thessaloniki
Priority to US12/429,832 priority Critical patent/US20100022631A1/en
Assigned to HADASIT MEDICAL RESEARCH SERVICES & DEVELOPMENT LTD. reassignment HADASIT MEDICAL RESEARCH SERVICES & DEVELOPMENT LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ILAN, YARON
Assigned to ARISTOTLE UNIVERSITY OF THESSALONIKI reassignment ARISTOTLE UNIVERSITY OF THESSALONIKI ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GRIGORIADIS, NICHOLAS, POUTACHIDIS, THEOFILOS
Assigned to YISSUM RESEARCH DEVELOPMENT COMPANY OF THE HEBREW UNIVERSITY OF JERUSALEM reassignment YISSUM RESEARCH DEVELOPMENT COMPANY OF THE HEBREW UNIVERSITY OF JERUSALEM ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AVRAHAM, YOSEFA, BERRY, ELLIOT, DAGON, YOSSI, MAGEN, IDDO, MECHOULAM, RAPHAEL
Publication of US20100022631A1 publication Critical patent/US20100022631A1/en
Priority to US13/162,160 priority patent/US10166202B2/en
Assigned to YISSUM RESEARCH DEVELOPMENT COMPANY OF THE HEBREW UNIVERSITY OF JERUSALEM LTD reassignment YISSUM RESEARCH DEVELOPMENT COMPANY OF THE HEBREW UNIVERSITY OF JERUSALEM LTD CORRECTIVE ASSIGNMENT TO CORRECT THE THE ASSIGNEE'S NAME PREVIOUSLY RECORDED AT REEL: 023339 FRAME: 0193. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: AVRAHAM, YOSEFA, BERRY, ELLIOT, DAGON, YOSSI, MAGEN, IDDO, MECHOULAM, RAPHAEL
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/075Ethers or acetals
    • A61K31/085Ethers or acetals having an ether linkage to aromatic ring nuclear carbon
    • A61K31/09Ethers or acetals having an ether linkage to aromatic ring nuclear carbon having two or more such linkages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/23Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms
    • A61K31/232Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms having three or more double bonds, e.g. etretinate
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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

Definitions

  • the present invention relates to a composition and methods for the treatment or prevention of hepatic encephalopathy and liver cirrhosis.
  • Cirrhosis is a consequence of acute and chronic liver disease characterized by replacement of liver tissue by fibrotic scar tissue as well as regenerative nodules, leading to progressive loss of liver function. Cirrhosis is most commonly caused by alcoholism, hepatitis C, toxins and fatty liver but has many other possible causes.
  • HE hepatic encephalopathy
  • cerebral dysfunction is heterogeneous ranging from mild neuropsychiatric and psychomotor dysfunction, impaired memory, increased reaction time, sensory abnormalities and poor concentration to severe features such as confusion, stupor, coma and eventually death.
  • Hepatic encephalopathy is caused by disorders affecting the liver including disorders that reduce liver function (such as cirrhosis or hepatitis) and conditions where there is impaired blood circulation in the liver.
  • liver failure induces impaired glucose oxidative pathways and increased lactate synthesis in the brain which results in energy failure.
  • hypoglycemia and hypoxia are also major contributors to the energy failure seen in hepatic encephalopathy.
  • ammonia is considered to play a major role in the pathogenesis of the neuropsychiatric disturbances observed in hepatic encephalopathy.
  • the liver is the major organ for detoxifying ammonia. When the liver fulls the body is incapable of efficiently converting ammonia to urea or glutamine, resulting in systemic hyperammonemia including the brain.
  • the brain lacks an effective urea cycle and therefore relies entirely on glutamine synthesis for the removal of blood-borne ammonia. Since glutamine synthetase is dependent on an adequate level of ATP to amidate glutamate to glutamine, ammonia intoxication results in depletion of brain ATP resources and eventually cell death (Ott et al., 2005; Hardie, 2004). Finally, decreased glucose utilization in the brain may be compensated by mobilization of amino acids to provide carbon skeletons as substrates for energy metabolism. Yet, attempts to balance energy failure at the expense of cerebral proteins may end in destructive brain proteolysis (Hardie and Carling, 1997).
  • AMP-activated protein kinase is an evolutionarily conserved metabolic master switch. AMPK is allosterically activated by 5′-AMP, which accumulates following ATP hydrolysis. Conversely, high ATP antagonizes the activating effects of 5′-AMP on AMPK. AMP binding to AMPK leads to activation of the enzyme by inducing a conformational change exposing threonine-172 in the catalytic domain, which undergoes phosphorylation by an upstream AMPK kinase (AMPKK) (Hawley et al., 1996).
  • AMPKK upstream AMPK kinase
  • Hypothalamic AMPK controls energy balance via regulation of food intake, body weight and glucose and lipid homeostasis (Dagon et al., 2005; Pagotto et al., 2005).
  • Hippocampal AMPK controls cognitive function via regulation of neurogenesis and neuroapoptosis (Dagon et al, 2005).
  • the cannabinoid (CB) system consists of two receptor subtypes.
  • the CB-1 receptors are predominantly found in the brain, while the CB-2 receptors are mostly found in the peripheral tissue (Matsuda, et al., 1990).
  • the main endogenous endocannabinoids are small molecules derived from membrane arachidonic acid, such as anandamide(arachidonoylethanolamide) and 2-arachidonoylglycerol (2-AG) (Iversen, 2000; Berry et al., 2002).
  • D9-tetrahydrocannabinol (THC), the major psychoactive constituent of the Cannabis plant, is a cannabinoid agonist which produces a myriad of complex pharmacological effects (Baker et al., 2003; Avraham et al., 2006). It is now recognized that most of the central effects of endogenous as well as exogenous cannabinoids are mediated through the CB-1 receptor, a family of G-protein-coupled receptors. Cerebral CB-1 receptors are part of the complex mechanisms involved in the control of energy balance via regulation of food intake and body weight (Teixeira-Clerc et al., 2006). The endocannabinoid system has also been demonstrated to exert neuroprotective effects in several types of cerebral insults via regulation of motor control, cognition, emotional responses, motivated behavior and homeostasis (Julien et al., 2005).
  • the endocannabinoid system was shown to have an important role in the pathogenesis of hepatic encephalopathy. Modulation of this system, either by specific antagonists to the CB1 cannabinoid receptor, or by agonists specific for the CB2 receptor, such as HU-308 was shown to be effective (Avraham et al., 2006).
  • the present invention is based on the surprising finding that D9-tetrahydrocannabinol (THC) is effective in the treatment of hepatic encephalopathy.
  • THC D9-tetrahydrocannabinol
  • This finding is surprising in view of the fact that THC was previously known to have about equal affinity both to the CB1 and the CB2 receptors and the above-mentioned Avraham et al, 2006 publication teaches that modulation of the endocannabinoid system is effected either by specific antagonists to the CB1 cannabinoid receptor or by agonists specific for the CB2 receptor.
  • the present invention is further based on the findings that cannabidiol (CBD) and capsaicin are effective in the treatment of hepatic encephalopathy. This finding is also surprising since cannabidiol does not exert its physiological activity through neither of the CB1 or the CB2 receptors while capsaicin is known to act through the vanilloid receptors subtype 1.
  • CBD cannabidiol
  • the present invention relates to a compound selected from D9-tetrahydrocannabinol (THC), cannabidiol or capsaicin, comprising said compound, for prevention, treatment, or both, of hepatic encephalopathy.
  • THC D9-tetrahydrocannabinol
  • cannabidiol cannabidiol
  • capsaicin comprising said compound, for prevention, treatment, or both, of hepatic encephalopathy.
  • the present invention also relates to pharmaceutical composition
  • a compound selected from D9-tetrahydrocannabinol (THC), cannabidiol or capsaicin for prevention, treatment, or both, of hepatic encephalopathy.
  • a method for prevention, treatment, or both, of hepatic encephalopathy comprising administering to a subject in need an effective amount of a compound selected from the group consisting of D9-tetrahydrocannabinol, cannabidiol and capsaicin, is provided.
  • hepatic encephalopathy in the context of the invention, and in accordance with the World Congress of Gastroenterology 1998 in Vienna, refers to all subclasses of the disease as follows: Type A (acute), hepatic encephalopathy associated with acute liver failure; type B (bypass), caused by portal-systemic shunting without associated intrinsic liver disease; and type C (cirrhosis), occurring in patients with cirrhosis.
  • hepatic encephalopathy refers to all durations and characteristics of hepatic encephalopathy and includes episodic, persistent and minimal.
  • minimal encephalopathy refers to patients with cirrhosis who do not demonstrate clinically overt cognitive dysfunction, but who show a cognitive impairment on neuropsychological studies.
  • the evaluation of severity of persistent hepatic encephalopathy is based on the West Haven Criteria for semi-quantitative grading of mental status, referring to the level of impairment of autonomy, changes in consciousness, intellectual function, behavior, and the dependence on therapy, and includes: Grade 1—trivial lack of awareness; euphoria or anxiety; shortened attention span; impaired performance of addition. Grade 2—lethargy or apathy; minimal disorientation for time or place; subtle personality change; inappropriate behavior; impaired performance of subtraction. Grade 3—somnolence to semistupor, but responsive to verbal stimuli; confusion; gross disorientation. Grade 4—Coma (unresponsive to verbal or noxious stimuli).
  • treatment in the context of the present invention refers to at least one of the following: decrease in the severity of at least one undesired side effect associated with the disease; improvement in the overall cognitive function of the treated subject; delay in the progression from one disease stage to the other; shortening the length of an hepatic encephalopathy episode and lengthening the period between episodes.
  • treatment is also meant to refer to preventive or prophylactic treatment—meaning that a person known to have liver dysfunction or to be at risk for developing liver dysfunction (for example, due to hepatitis C) is administered with THC, cannabidiol or capsaicin, even before manifestation of hepatic encephalopathy in order to prevent its occurrence.
  • THC or “D9-tetrahydrocannabinol” are used herein interchangeably for the compound ( ⁇ )-(6aR,10aR)-6,6,9-trimethyl-3-pentyl-6a,7,8,10a-tetrahydro-6H-benzo[c]chromen-1-ol.
  • This substance may be isolated from the natural source (cannabis), for example, in accordance with the method in Gaoni and Mechoulam (1964) or may be synthetically produced such as dronabinol, which is available as a prescription drug (under the trade name MarinolTM of Unimed Pharmaceuticals, Inc.)
  • the present invention also relates to a compound selected from capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 or cannabidiol for prevention, treatment, or both, of liver cirrhosis.
  • the invention further relates to the use of capsaicin, cannabidiol (CBD), 2-arachidonoylglycerol or HU-308 for the preparation of a medicament for prevention, treatment, or both, of liver cirrhosis.
  • CBD cannabidiol
  • 2-arachidonoylglycerol or HU-308 for the preparation of a medicament for prevention, treatment, or both, of liver cirrhosis.
  • the invention concerns a method for the prevention, treatment, or both, of liver cirrhosis comprising administering to a subject a therapeutically effective amount of capsaicin, cannabidiol, 2-arachidonoylglycerol or HU-308.
  • liver cirrhosis refers to any stage in the development of the pathological condition, from very initial development of fibrotic scar tissue to full-blown liver cirrhosis.
  • diseases or conditions that are known to lead to liver cirrhosis are, but are not limited to: alcoholic liver disease, chronic viral hepatitis (Type B and C), chronic bile duct blockage, metabolic diseases resulting in abnormal storage of copper (Wilson's disease) or iron (Hemochromatosis).
  • Cirrhosis may also be caused by exposure to drugs and toxins, by autoimmune processes such as autoimmune hepatitis, by inherited diseases such as cystic fibrosis and alpha antitrypsin deficiency, and by obesity (so called “fatty liver” or nonalcoholic steatohepatitis). Furthermore, severe reactions to prescription drugs, prolonged exposure to environmental toxins such as arsenic, the parasitic infection schistosomiasis, and repeated bouts of heart failure with liver congestion can all lead to cirrhosis.
  • the treatment may be initiated when a disease is established to stop or slow disease progression.
  • a disease e.g. hepatitis, excessive consumption of alcohol and obesity
  • capsaicin, cannabidiol, 2-AG or HU-308 may be given in a preventive prophylactic manner to prevent or delay the onset of cirrhosis.
  • the level of significance of differences between treatment groups is designated with one, two or three asterisks(s).
  • FIG. 1 shows D9-tetrahydrocannabinol (THC)-induced AMPK activation following thioacetamide (TAA)-induced liver failure in mice.
  • THC D9-tetrahydrocannabinol
  • TAA thioacetamide
  • FIGS. 2A-E show that THC activates AMPK and improves impaired brain function in TAA-induced liver failure in mice.
  • Mice were treated with TAA for 5 days, then 0.1 mM THC was administrated daily for 5 days.
  • 2 A Hippocampal AMPK expression and phosphorylation on Thr 172 were analyzed by immunoblotting. P-AMPK, phosphorylated AMPK.
  • 2 B Mice were treated as above and performance in an eight arm maze was measured every day after the THC treatment. AUC, area under the curve.
  • 2 C Activity score.
  • 2 D Neurological score under the same conditions.
  • Catecholamines levels were analyzed by HPLC. DA, dopamine.
  • FIGS. 3A-F show the effect of AICAR and THC treatment on hepatic failure.
  • Mice were treated with TAA or saline and then with 0.5 mM AICAR or 0.1 mg/l THC. Blood plasma was obtained for liver functions analysis.
  • 3 A Ammonia
  • 3 B Bilirubin
  • 3 C alanine transaminase
  • AST aspartate aminotransferase
  • 3 E gamma-glutamyltransferase
  • GTT gamma-glutamyltransferase
  • FIGS. 4A-F depict histopathological changes in the liver after treatment with TAA.
  • FIGS. 5A-B show TAA effect on ( 5 A) alanine transaminase (ALT) and ( 5 B) aspartate aminotransferase (AST) in mice treated with different cannabinoid receptor ligands.
  • FIGS. 6A-B show effect of 2-arachidonoylglycerol (2-AG) and SR141716A on blood ALT ( 6 A) and AST levels ( 6 B) in TAA treated CB2-KO mice.
  • FIGS. 7A-B show that capsaicin significantly reduces both the ALT ( 7 A) and the AST ( 7 B) level in TAA treated mice.
  • FIGS. 8A-F depict glial cell staining at the area of hippocampus in na ⁇ ve animals ( 8 A), and astrogliosis following TAA administration ( 8 B-F).
  • GFAP a marker for glial cells
  • FIGS. 9A-C show that capsaicin significantly improves TAA-induced impaired cognitive function ( 9 A), poor activity performances ( 9 B) and reduced neurological score ( 9 C).
  • FIGS. 10A-B depict the effect of 2-AG treatment of chronic liver failure induced by bile duct ligation (BDL) on cognitive impairment ( 10 A) and motor impairment ( 10 B) relative to Sham operated animals.
  • AUC area under the curve.
  • FIGS. 11A-B depict the effect of cannabidiol (CBD) treatment of chronic liver failure induced by bile duct ligation (BDL) on cognitive impairment ( 11 A) and motor impairment ( 11 ) relative to Sham operated animals.
  • CBD cannabidiol
  • FIGS. 11A-B depict the effect of cannabidiol (CBD) treatment of chronic liver failure induced by bile duct ligation (BDL) on cognitive impairment ( 11 A) and motor impairment ( 11 ) relative to Sham operated animals.
  • AUC area under the curve.
  • FIGS. 12A-B depict the effect of cannabidiol (CBD) treatment of chronic liver failure induced by bile duct ligation (BDL) on IL-1 ⁇ mRNA level in the hippocampus relative to Sham operated animals.
  • 12 A RT-PCR gel separation; 12 B quantification of measurements done on the gel depicted in 12 A.
  • L19 ribosomal protein commonly used as invariant control gene; AUC, area under the curve.
  • FIG. 13 shows the effect of cannabidiol (CBD) treatment of chronic liver failure induced by bile duct ligation (BDL) on oxidative stress in the liver.
  • CBD cannabidiol
  • BDL bile duct ligation
  • AMPK is potently activated in murine models of hepatic encephalopathy. This correlates with the observed hyperammonia and hypoglycemia—two major causes of cerebral energy depletion. Nonetheless, as found in acute hepatotoxicity (caused by TAA), this response decreases with time, and eventually reaches the same level as that of the chronic stress induced by bile duct ligation. Such a cerebral adaptation response fulls to meet the intact brain energy requirements and may be augmented by pharmacological means (AICAR).
  • AICAR pharmacological means
  • AMPK pharmacological activation of AMPK might provide a new strategy for the management of hepatic encephalopathy.
  • unselective drugs such as AICAR, which activate AMPK under normal as well as under stress conditions, are not suitable for clinical use.
  • THC the main active constituent of marijuana, has been repeatedly demonstrated to cause brain dysfunction and neurotoxicity (Mishima et al., 2001). This finding is in line with our observations disclosed herein below of its ability to stimulate AMPK.
  • these studies have used high dosage of THC (1-15 mg/kg), quantities that we found necessary for AMPK activation under normal circumstances.
  • THC could be suitable as a selective agent that could function as a “stress specific drug” by activating AMPK only under pathological conditions.
  • THC which has about equal affinity for the CB1 and CB2 receptor
  • THC transient receptor potential 1 TRPV1
  • capsaicin suggested by Di Marzo et al (1998) to interact with the endocannabinoid system, acts on neural cells via vanilloid receptors subtype 1 (VR1, also known as transient receptor potential 1 TRPV1), a non-selective cation channel, which can be blocked by capsazepine.
  • VR1 vanilloid receptors subtype 1
  • TRPV1 transient receptor potential 1
  • a second compound tested herein to treat the animals is cannabidiol, an active ingredient of Cannabis Sativa devoid of adverse effects related to the CB1 receptor owing to its CB1-independent mechanism of action.
  • Cannabidiol is also a very potent anti-inflammatory agent. It is a finding of the present invention that cannabidiol improves impaired brain and liver function in experimental hepatic encephalopathy in animal models. Furthermore, two additional cannabinoids, 2-AG and HU-308 were shown herein to positively affect liver function in experimental hepatic encephalopathy in animal models.
  • the present invention thus provides a compound selected from D9-tetrahydrocannabinol (THC), cannabidiol and capsaicin for prevention, treatment, or both of hepatic encephalopathy.
  • THC D9-tetrahydrocannabinol
  • cannabidiol cannabidiol
  • capsaicin for prevention, treatment, or both of hepatic encephalopathy.
  • the compound is D9-tetrahydrocannabinol. In another preferred embodiment the compound is cannabidiol. In still another preferred embodiment the compound is capsaicin.
  • the compound may be formulated in any suitable form for administration, preferably in an oral, parenteral, sublingual or intranasal dosage form.
  • D9-tetrahydrocannabinol, cannabidiol and capsaicin are intended for prevention and/or treatment of all subclasses of hepatic encephalopathy as described above, i.e. Type A, Type B or Type C, preferably type A or type C.
  • the present invention further provides a pharmaceutical composition for prevention, treatment, or both, of hepatic encephalopathy comprising a compound selected from D9-tetrahydrocannabinol, cannabidiol and capsaicin and a pharmaceutically acceptable carrier.
  • the present invention also concerns a method for prevention, treatment, or both, of hepatic encephalopathy comprising administering to a subject in need a therapeutically effective amount of a compound selected from D9-tetrahydrocannabinol (THC), cannabidiol and capsaicin.
  • THC D9-tetrahydrocannabinol
  • cannabidiol cannabidiol
  • the present invention relates to a compound selected from capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 or cannabidiol for prevention, treatment, or both, of liver cirrhosis.
  • the invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound, selected from capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 or cannabidiol for prevention, treatment, or both, of liver cirrhosis
  • the compound is 2-arachidonoylglycerol. In another preferred embodiment the compound is HU-308. In still another preferred embodiment the compound is capsaicin. In yet another preferred embodiment the compound is cannabidiol.
  • prevention of liver cirrhosis refers herein to preventing or slowing the deterioration of any damage caused to the liver tissue, such as the accumulation of fibrotic scar tissue, by factors known to cause cirrhosis such as, but not limited to, alcoholic liver disease, chronic viral hepatitis type C, chronic viral hepatitis type B, chronic bile duct blockage, Wilson's disease, hemochromatosis, exposures to drug and toxins, autoimmune hepatitis, cystic fibrosis, alpha antitrypsin deficiency, obesity or schistosomiasis.
  • liver cirrhosis prevention of the development of liver cirrhosis can be achieved by treating subjects in need, such as alcoholics, people infected with hepatitis C and obese people, at very early stages of their disease or condition, even before appearance of physical symptoms of liver cirrhosis.
  • the present invention also concerns a method for prevention, treatment, or both, of liver cirrhosis, comprising administering to a subject in need a therapeutically effective amount of a compound selected from capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 or cannabidiol.
  • the present invention further provides a compound selected from cannabidiol (CBD) or capsaicin for prevention, treatment, or both, of hepatic encephalopathy or liver cirrhosis.
  • CBD cannabidiol
  • the invention further provides a pharmaceutical composition for prevention, treatment, or both, of hepatic encephalopathy or liver cirrhosis, comprising cannabidiol or capsaicin and a pharmaceutically acceptable carrier.
  • THC, SR141716A, SR144528 and CBD were provided by Prof. Raphael Mechoulam (Faculty of Medicine and Department of Pharmacology, Hebrew University of Jerusalem).
  • Hepatotoxin thioacetamide (TAA) and capsaicin were obtained from Sigma-Aldrich (Rehovot, Israel).
  • 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) was obtained from Toronto Research Chemicals (TRC).
  • HU-308 was synthesized as described in Hanus et al. (1999).
  • mice Eight- to 10-week old female Sabra mice (29-32 g), obtained from the animal facility of the Hebrew University, Israel, were assigned at random to different groups of 10 mice per cage and were used in all experiments. All cages contained wood-chip bedding and were placed in a temperature-controlled room at 22° C., on a 12 h light/dark cycle (lights on at 07.00 a.m.). The mice had free access to water 24 h a day. The food provided was Purina chow, the animals were maintained in the animal facility (SPF unit) of the Hebrew University Hadassah Medical School, Jerusalem. CB-2 KO mice were provided by Prof. Zimmer, Institute of Molecular Psychiatry, University of Bonn, Germany.
  • mice were sacrificed after treatment by decapitation between 10.00-12.00 a.m. Brains were rapidly removed and were dissected out and kept at ⁇ 70° C.
  • Bile duct ligation (iiia) Bile duct ligation. A midline incision was made under general anesthesia. The common bile duct was localized, doubly ligated, and cut between these two ligatures. In sham animals, a midline incision was performed, but with BDL.
  • TAA TAA.
  • a single dose of 200 mg kg-1 of TAA was injected by the intraperitoneal route (i.p.). 24 hours after injection all animals (including control) were injected (s. c) with 0.5 ml solution of 0.45% NaCl, 5% dextrose and 0.2% KCl in order to prevent hypovolemia, hypokalemia and hypoglycemia. The mice were intermittently exposed to infrared light in order to prevent hypothermia.
  • THC was administered i.p. either alone or with SR141716A on day 6 after TAA administration. Mice were sacrificed 1 h post treatment and analyzed for AMPK level. For the behavioral tests which started on day 6 after TAA administration, THC was administered i.p. during days 6-10. Neurological score, activity and cognitive function were analyzed during these days.
  • Non-specific binding in a Western blot analysis was prevented by immersing the membranes in blocking buffer (5% nonfat dry milk in Tris-buffer saline-Tween 20 (TBS-T)), for 2 h at room temperature. The membranes were then exposed to the indicated antibodies diluted 1:1000 for 1 h at room temperature.
  • Anti-AMPK and phospho-AMPK antibodies were obtained from Cell Signaling.
  • Anti-protein kinase B (AKT) was obtained from Upstate.
  • Anti-actin was from Santa Cruz Biotechnology Inc. (Santa Cruz, Calif.).
  • the blots were rinsed in TBS-T and then incubated with horseradish peroxidase-conjugated goat anti-mouse antibodies (1:10,000; Santa Cruz Biotechnology Inc., Santa Cruz, Calif.) for 1 h at room temperature. Antibody-antigen complexes were visualized by detecting enhanced chemiluminescence with X-ray film.
  • Primers specific for CB1 were GGAGAACATCCAGTGTGGGG [SEQ ID NO: 1] and CATTGGGGCTGTCTTTACGG [SEQ ID NO: 2], for CB2 GGGTCCTCTCAGCATTGATTT [SEQ ID NO: 3], and GTTAACAAGGCACAGCATGGAAC [SEQ ID NO: 4], and for actin CAG CTTCTTTGCAGCTCCTT [SEQ ID NO: 5] and TCACCCACATAGGAGTCCT [SEQ ID NO: 6]. All primers were synthesized by Danyel Biotech, Israel.
  • Neurological function was assessed by a 10 point scale based on reflexes and task performance (Chen et al., 1996): exit from a circle 1 meter in diameter in less than 1 minute, seeking, walking a straight line, startle reflex, grasping reflex, righting reflex, placing reflex, corneal reflex, maintaining balance on a beam 3, 2 and 1 cm in width, climbing onto a square and a round pole. For each task failed or abnormal reflex reaction a score of 1 was assigned. Thus, a higher score indicates poorer neurological function. The neurological score was assessed one day after TAA induction (day 2). The mice were then divided between treatment groups so that each group had a similar baseline neurological score after TAA induction. The post-treatment neurological score was assessed one day after administration of the agonist or the antagonist or the vehicle (day 3).
  • (viii) Activity The activity test was performed on day 4, since in the first 3 days after TAA injection almost no motor activity was observed.
  • One of two methods was utilized: a) an activity apparatus, which consists of a cylindrical chamber (60 cm in diameter) with crossing infrared beams. Locomotor activity was recorded by a counter (attached to the apparatus), that counts the number of beam crossings made by the mice at one-minute intervals. Activity of two mice was measured simultaneously for a five-minute period. Two mice were tested together to lower stress to the minimum. Activity is presented as the mean number of beam crossings in 5 minutes.
  • THC treatment showed a biphasic effect (Sulcova et al., 1998). While low levels of THC (less than 0.1 mg/kg) reduced the level of activated AMPK, higher concentrations exhibited a dose dependent elevation in activated enzyme, reaching a significant activation of AMPK ( FIG. 1 ).
  • THC was tested in TAA treated mice. In this instance THC also demonstrated a biphasic effect.
  • THC Activates AMPK and Improves Impaired Brain Function in Experimental Hepatic Encephalopathy
  • THC which has about equal affinity for the CB1 and CB2 receptor
  • THC which has about equal affinity for the CB1 and CB2 receptor
  • capsaicin suggested by Di Marzo et al (1998) to interact with the endocannabinoid system, acts on neural cells via vanilloid receptors subtype 1 (VR1, also known as transient receptor potential 1 TRPV1), a non-selective cation channel, which can be blocked by capsazepine.
  • VR1 vanilloid receptors subtype 1
  • TRPV1 transient receptor potential 1
  • FIGS. 4A-F depict varying degrees of necrosis which were semi-quantitated as histopathological indices 1-4.
  • TAA treated mice demonstrated increased levels of alanine transaminase (ALT) ( FIG. 5A ) (see also FIG. 3C ) and aspartate aminotransferase (AST) ( FIG. 5B ) (see also FIG. 3D ); treatment of the TAA treated mice with 2-AG—a CB1 agonist, SR141716A—a CB1 antagonist, HU-308—a CB2 agonist, and SR144528—a CB2 antagonist, all significantly reduced ALT and AST levels. Moreover, 2-AG did not counteract the effect of SR141716A or SR144528, and HU-308 did not counteract the effect of SR144528. Thus, the results imply that the agonists/antagonists did not convey their effect specifically through the CB1 or CB2 receptors.
  • CB2 Agonist, but not CB1 Antagonist Treatment Improves Markers of Hepatic Function in CB2-KO Mice
  • FIGS. 7A-B show that indeed, capsaicin significantly reduces both the ALT and the AST level in TAA treated mice.
  • the effect of capsaicin is specifically affected through the VR1 receptor as evidenced by the abolishment of the effect of capsaicin by the VR1 antagonist capsazepine.
  • endocannabioid agonist and antagonist and capsaicin treatment affect the important aspect of hepatic encephalopathy pathology—astrogliosis—TAA treated animals were treated with these compounds and hippocampus was stained for glial cells in na ⁇ ve animals ( FIG. 8A ) and after treatment ( FIGS. 8B-F ).
  • FIG. 8B hepatic encephalopathy induced intensive glial fibrillary acidic protein (GFAP, a marker for glial cells) staining intensity and increased process complexity, i.e. more processes and increased branching ( FIG. 8B ) as compared with na ⁇ ve animals ( FIG. 8 A).
  • FIGS. 10A-B 2-AG effectively reversed cognitive impairments secondary to biliary cirrhosis in mice ( FIG. 10A ), but failed to reverse the motor impairments ( FIG. 10B ) which are also typical to this disorder.
  • Cannabidiol Improves Impaired Brain and Liver Function in Experimental Hepatic Encephalopathy
  • Cannabidiol an active ingredient of Cannabis Sativa devoid of adverse effects related to the CB1 receptor owing to its CB1-independent mechanism of action.
  • Cannabidiol is also a very potent anti-inflammatory agent.
  • BDL bile duct ligation
  • mice Two weeks post-surgery, the cognitive and the motor function of the mice were evaluated. Mice were decapitated 3 weeks post-surgery and their hippocampi were taken for analysis of IL-1b mRNA level by RT-PCR. The results clearly show that cognitive function ( FIG. 11A ) and motor activity ( FIG. 11B ) are impaired by BDL after 2 weeks and is restored by cannabidiol. Also, IL-1 ⁇ mRNA level (normalized to ribosomal protein L19 mRNA levels; a commonly used invariant control gene) in the hippocampus is elevated following BDL and is restored by cannabidiol ( FIGS. 12A-B ).
  • Oxidative stress in liver tissue due to chronic liver failure induced by BDL was assessed by measuring malondialdehyde, a well accepted biomarker for oxidative stress. As can be seen in FIG. 13 , oxidative stress is elevated following BDL and is restored by cannabidiol. As oxidative stress is commonly known to be involved in the development of cirrhosis (Ara et al., 2005) and treatment with cannabidiol reduces the oxidative stress, one can deduce that this treatment will prevent or slow down the development of cirrhosis.
  • bile duct ligation induced cognitive and motor deficits and increased oxidative stress in the liver, which were reversed by cannabidiol.
  • cannabidiol In the hippocampus, which is responsible for learning and memory, there was an up-regulation of IL-1b mRNA following BDL, which was also reversed by cannabidiol, suggesting causal relationship between an inflammatory response in this region and impaired learning.

Abstract

The compounds D9-tetrahydrocannabinol (THC), cannabidiol (CBD) and capsaicin are useful for prevention, treatment, or both, of hepatic encephalopathy. The compounds capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 and cannabidiol are useful for prevention, treatment, or both, of liver cirrhosis.

Description

    FIELD OF THE INVENTION
  • The present invention relates to a composition and methods for the treatment or prevention of hepatic encephalopathy and liver cirrhosis.
  • BACKGROUND OF THE INVENTION
  • Cirrhosis is a consequence of acute and chronic liver disease characterized by replacement of liver tissue by fibrotic scar tissue as well as regenerative nodules, leading to progressive loss of liver function. Cirrhosis is most commonly caused by alcoholism, hepatitis C, toxins and fatty liver but has many other possible causes.
  • Ascites (fluid retention in the abdominal cavity) is the most common complication of cirrhosis and is associated with a poor quality of life, increased risk of infection, and a poor long-term outcome. Other potentially life-threatening complications are hepatic encephalopathy and bleeding from esophageal varices. Today, cirrhosis is generally irreversible once it occurs, and treatment generally focuses on preventing progression and complications. In advanced stages of cirrhosis the only option is a liver transplant.
  • Modern medicine defines hepatic encephalopathy (HE) as a neuropsychiatric syndrome, which is associated with acute or chronic liver dysfunction and has quantitatively and qualitatively distinct features relating to its severity. In cirrhosis, cerebral dysfunction is heterogeneous ranging from mild neuropsychiatric and psychomotor dysfunction, impaired memory, increased reaction time, sensory abnormalities and poor concentration to severe features such as confusion, stupor, coma and eventually death.
  • Hepatic encephalopathy is caused by disorders affecting the liver including disorders that reduce liver function (such as cirrhosis or hepatitis) and conditions where there is impaired blood circulation in the liver.
  • While the symptoms of hepatic encephalopathy are well documented, its pathogenesis is not clear yet and a number of possible scenarios have been suggested. First, liver failure induces impaired glucose oxidative pathways and increased lactate synthesis in the brain which results in energy failure. Second, hypoglycemia and hypoxia are also major contributors to the energy failure seen in hepatic encephalopathy. Third, ammonia is considered to play a major role in the pathogenesis of the neuropsychiatric disturbances observed in hepatic encephalopathy. The liver is the major organ for detoxifying ammonia. When the liver fulls the body is incapable of efficiently converting ammonia to urea or glutamine, resulting in systemic hyperammonemia including the brain. Unlike the liver, the brain lacks an effective urea cycle and therefore relies entirely on glutamine synthesis for the removal of blood-borne ammonia. Since glutamine synthetase is dependent on an adequate level of ATP to amidate glutamate to glutamine, ammonia intoxication results in depletion of brain ATP resources and eventually cell death (Ott et al., 2005; Hardie, 2004). Finally, decreased glucose utilization in the brain may be compensated by mobilization of amino acids to provide carbon skeletons as substrates for energy metabolism. Yet, attempts to balance energy failure at the expense of cerebral proteins may end in destructive brain proteolysis (Hardie and Carling, 1997).
  • However, other factors such as an inflammatory response and astrogliosis in the brain are also implicated in hepatic encephalopathy.
  • The AMP-activated protein kinase (AMPK) is an evolutionarily conserved metabolic master switch. AMPK is allosterically activated by 5′-AMP, which accumulates following ATP hydrolysis. Conversely, high ATP antagonizes the activating effects of 5′-AMP on AMPK. AMP binding to AMPK leads to activation of the enzyme by inducing a conformational change exposing threonine-172 in the catalytic domain, which undergoes phosphorylation by an upstream AMPK kinase (AMPKK) (Hawley et al., 1996).
  • Once activated, it switches on catabolic pathways (such as fatty acid oxidation and glycolysis) and switches off ATP-consuming pathways (such as lipogenesis) both by short-term effect on phosphorylation of regulatory proteins and by long-term effect on gene expression (Foretz et al., 2006). Stresses such as nutrient depletion, hypoxia, heat shock, metabolic poisoning and exercise, all activate AMPK by their effect on the ratio of 5′-AMP to ATP. AMPK, in turn, phosphorylates multiple targets, which switch off anabolic pathways and stimulate catabolic ones. AMPK was recently recognized as a key regulator of whole body energy metabolism (Minokoshi et al., 2004). Cerebral AMPK responds to integration of nutritional and hormonal input. Hypothalamic AMPK controls energy balance via regulation of food intake, body weight and glucose and lipid homeostasis (Dagon et al., 2005; Pagotto et al., 2005). Hippocampal AMPK controls cognitive function via regulation of neurogenesis and neuroapoptosis (Dagon et al, 2005).
  • The cannabinoid (CB) system consists of two receptor subtypes. The CB-1 receptors are predominantly found in the brain, while the CB-2 receptors are mostly found in the peripheral tissue (Matsuda, et al., 1990). The main endogenous endocannabinoids are small molecules derived from membrane arachidonic acid, such as anandamide(arachidonoylethanolamide) and 2-arachidonoylglycerol (2-AG) (Iversen, 2000; Berry et al., 2002). D9-tetrahydrocannabinol (THC), the major psychoactive constituent of the Cannabis plant, is a cannabinoid agonist which produces a myriad of complex pharmacological effects (Baker et al., 2003; Avraham et al., 2006). It is now recognized that most of the central effects of endogenous as well as exogenous cannabinoids are mediated through the CB-1 receptor, a family of G-protein-coupled receptors. Cerebral CB-1 receptors are part of the complex mechanisms involved in the control of energy balance via regulation of food intake and body weight (Teixeira-Clerc et al., 2006). The endocannabinoid system has also been demonstrated to exert neuroprotective effects in several types of cerebral insults via regulation of motor control, cognition, emotional responses, motivated behavior and homeostasis (Julien et al., 2005).
  • The endocannabinoid system was shown to have an important role in the pathogenesis of hepatic encephalopathy. Modulation of this system, either by specific antagonists to the CB1 cannabinoid receptor, or by agonists specific for the CB2 receptor, such as HU-308 was shown to be effective (Avraham et al., 2006).
  • SUMMARY OF THE INVENTION
  • The present invention is based on the surprising finding that D9-tetrahydrocannabinol (THC) is effective in the treatment of hepatic encephalopathy. This finding is surprising in view of the fact that THC was previously known to have about equal affinity both to the CB1 and the CB2 receptors and the above-mentioned Avraham et al, 2006 publication teaches that modulation of the endocannabinoid system is effected either by specific antagonists to the CB1 cannabinoid receptor or by agonists specific for the CB2 receptor.
  • The present invention is further based on the findings that cannabidiol (CBD) and capsaicin are effective in the treatment of hepatic encephalopathy. This finding is also surprising since cannabidiol does not exert its physiological activity through neither of the CB1 or the CB2 receptors while capsaicin is known to act through the vanilloid receptors subtype 1.
  • Accordingly, the present invention relates to a compound selected from D9-tetrahydrocannabinol (THC), cannabidiol or capsaicin, comprising said compound, for prevention, treatment, or both, of hepatic encephalopathy.
  • The present invention also relates to pharmaceutical composition comprising a compound selected from D9-tetrahydrocannabinol (THC), cannabidiol or capsaicin, for prevention, treatment, or both, of hepatic encephalopathy.
  • Furthermore a method for prevention, treatment, or both, of hepatic encephalopathy comprising administering to a subject in need an effective amount of a compound selected from the group consisting of D9-tetrahydrocannabinol, cannabidiol and capsaicin, is provided.
  • The term “hepatic encephalopathy”, in the context of the invention, and in accordance with the World Congress of Gastroenterology 1998 in Vienna, refers to all subclasses of the disease as follows: Type A (acute), hepatic encephalopathy associated with acute liver failure; type B (bypass), caused by portal-systemic shunting without associated intrinsic liver disease; and type C (cirrhosis), occurring in patients with cirrhosis.
  • This term refers to all durations and characteristics of hepatic encephalopathy and includes episodic, persistent and minimal. The term “minimal encephalopathy” refers to patients with cirrhosis who do not demonstrate clinically overt cognitive dysfunction, but who show a cognitive impairment on neuropsychological studies.
  • The evaluation of severity of persistent hepatic encephalopathy is based on the West Haven Criteria for semi-quantitative grading of mental status, referring to the level of impairment of autonomy, changes in consciousness, intellectual function, behavior, and the dependence on therapy, and includes: Grade 1—trivial lack of awareness; euphoria or anxiety; shortened attention span; impaired performance of addition. Grade 2—lethargy or apathy; minimal disorientation for time or place; subtle personality change; inappropriate behavior; impaired performance of subtraction. Grade 3—somnolence to semistupor, but responsive to verbal stimuli; confusion; gross disorientation. Grade 4—Coma (unresponsive to verbal or noxious stimuli).
  • The term “treatment” in the context of the present invention refers to at least one of the following: decrease in the severity of at least one undesired side effect associated with the disease; improvement in the overall cognitive function of the treated subject; delay in the progression from one disease stage to the other; shortening the length of an hepatic encephalopathy episode and lengthening the period between episodes.
  • The term “treatment” is also meant to refer to preventive or prophylactic treatment—meaning that a person known to have liver dysfunction or to be at risk for developing liver dysfunction (for example, due to hepatitis C) is administered with THC, cannabidiol or capsaicin, even before manifestation of hepatic encephalopathy in order to prevent its occurrence.
  • The terms “THC” or “D9-tetrahydrocannabinol” are used herein interchangeably for the compound (−)-(6aR,10aR)-6,6,9-trimethyl-3-pentyl-6a,7,8,10a-tetrahydro-6H-benzo[c]chromen-1-ol. This substance may be isolated from the natural source (cannabis), for example, in accordance with the method in Gaoni and Mechoulam (1964) or may be synthetically produced such as dronabinol, which is available as a prescription drug (under the trade name Marinol™ of Unimed Pharmaceuticals, Inc.)
  • The present invention also relates to a compound selected from capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 or cannabidiol for prevention, treatment, or both, of liver cirrhosis.
  • The invention further relates to the use of capsaicin, cannabidiol (CBD), 2-arachidonoylglycerol or HU-308 for the preparation of a medicament for prevention, treatment, or both, of liver cirrhosis.
  • Furthermore the invention concerns a method for the prevention, treatment, or both, of liver cirrhosis comprising administering to a subject a therapeutically effective amount of capsaicin, cannabidiol, 2-arachidonoylglycerol or HU-308.
  • The term “liver cirrhosis” as used herein refers to any stage in the development of the pathological condition, from very initial development of fibrotic scar tissue to full-blown liver cirrhosis. Examples of diseases or conditions that are known to lead to liver cirrhosis are, but are not limited to: alcoholic liver disease, chronic viral hepatitis (Type B and C), chronic bile duct blockage, metabolic diseases resulting in abnormal storage of copper (Wilson's disease) or iron (Hemochromatosis). Cirrhosis may also be caused by exposure to drugs and toxins, by autoimmune processes such as autoimmune hepatitis, by inherited diseases such as cystic fibrosis and alpha antitrypsin deficiency, and by obesity (so called “fatty liver” or nonalcoholic steatohepatitis). Furthermore, severe reactions to prescription drugs, prolonged exposure to environmental toxins such as arsenic, the parasitic infection schistosomiasis, and repeated bouts of heart failure with liver congestion can all lead to cirrhosis.
  • The treatment may be initiated when a disease is established to stop or slow disease progression. Alternatively, as many of the conditions (e.g. hepatitis, excessive consumption of alcohol and obesity) are evident long before cirrhosis develops, often many years before, capsaicin, cannabidiol, 2-AG or HU-308 may be given in a preventive prophylactic manner to prevent or delay the onset of cirrhosis.
  • BRIEF DESCRIPTION OF THE FIGURES
  • The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fee.
  • In some of the following figures, the level of significance of differences between treatment groups is designated with one, two or three asterisks(s).
  • FIG. 1 shows D9-tetrahydrocannabinol (THC)-induced AMPK activation following thioacetamide (TAA)-induced liver failure in mice. Mice were administrated with saline or TAA. After 5 days, mice were treated with 0.1-10 mg/kg THC for 1 h. AMPK expression and phosphorylation on Thr172 were analyzed by immunoblotting. Black columns represent control group and the gray columns represents the TAA-treated group.
  • FIGS. 2A-E show that THC activates AMPK and improves impaired brain function in TAA-induced liver failure in mice. Mice were treated with TAA for 5 days, then 0.1 mM THC was administrated daily for 5 days. (2A) Hippocampal AMPK expression and phosphorylation on Thr 172 were analyzed by immunoblotting. P-AMPK, phosphorylated AMPK. (2B) Mice were treated as above and performance in an eight arm maze was measured every day after the THC treatment. AUC, area under the curve. (2C) Activity score. (2D) Neurological score under the same conditions. (2E) Catecholamines levels were analyzed by HPLC. DA, dopamine.
  • FIGS. 3A-F show the effect of AICAR and THC treatment on hepatic failure. Mice were treated with TAA or saline and then with 0.5 mM AICAR or 0.1 mg/l THC. Blood plasma was obtained for liver functions analysis. (3A) Ammonia; (3B) Bilirubin; (3C) alanine transaminase (ALT); (3D) aspartate aminotransferase (AST); (3E) gamma-glutamyltransferase (GGT); (3F) Glucose.
  • FIGS. 4A-F depict histopathological changes in the liver after treatment with TAA. 4A, normal mouse liver histology-score=0; 4B, mild centrilobular necrosis/apoptosis (score=1); FIG. 4C, Centrilobular coagulative necrosis (score=2); 4D, Central to central (bridging) necrosis (score=3); 4E, massive necrosis effacing liver architecture (score=4); 4F, higher magnification of liver centrilobular area (score=1).
  • FIGS. 5A-B show TAA effect on (5A) alanine transaminase (ALT) and (5B) aspartate aminotransferase (AST) in mice treated with different cannabinoid receptor ligands.
  • FIGS. 6A-B show effect of 2-arachidonoylglycerol (2-AG) and SR141716A on blood ALT (6A) and AST levels (6B) in TAA treated CB2-KO mice.
  • FIGS. 7A-B show that capsaicin significantly reduces both the ALT (7A) and the AST (7B) level in TAA treated mice.
  • FIGS. 8A-F depict glial cell staining at the area of hippocampus in naïve animals (8A), and astrogliosis following TAA administration (8B-F). Hepatic encephalopathy induced intensive glial fibrillary acidic protein (GFAP, a marker for glial cells) staining intensity and increased process complexity. These changes were minimal following capsaicin—(8C), CB1 antagonist—(8D) and CB2 agonist—(8E) treatment, whereas CB2 antagonist administration (8F) did not alter either the number or morphology of activated astroglia compared to untreated animals (8B).
  • FIGS. 9A-C show that capsaicin significantly improves TAA-induced impaired cognitive function (9A), poor activity performances (9B) and reduced neurological score (9C).
  • FIGS. 10A-B depict the effect of 2-AG treatment of chronic liver failure induced by bile duct ligation (BDL) on cognitive impairment (10A) and motor impairment (10B) relative to Sham operated animals. AUC, area under the curve.
  • FIGS. 11A-B depict the effect of cannabidiol (CBD) treatment of chronic liver failure induced by bile duct ligation (BDL) on cognitive impairment (11A) and motor impairment (11) relative to Sham operated animals. AUC, area under the curve.
  • FIGS. 12A-B depict the effect of cannabidiol (CBD) treatment of chronic liver failure induced by bile duct ligation (BDL) on IL-1β mRNA level in the hippocampus relative to Sham operated animals. 12A, RT-PCR gel separation; 12B quantification of measurements done on the gel depicted in 12A. L19, ribosomal protein commonly used as invariant control gene; AUC, area under the curve.
  • FIG. 13 shows the effect of cannabidiol (CBD) treatment of chronic liver failure induced by bile duct ligation (BDL) on oxidative stress in the liver. MDA, malondialdehyde.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Our study shows that AMPK is potently activated in murine models of hepatic encephalopathy. This correlates with the observed hyperammonia and hypoglycemia—two major causes of cerebral energy depletion. Nonetheless, as found in acute hepatotoxicity (caused by TAA), this response decreases with time, and eventually reaches the same level as that of the chronic stress induced by bile duct ligation. Such a cerebral adaptation response fulls to meet the intact brain energy requirements and may be augmented by pharmacological means (AICAR).
  • In light of this, pharmacological activation of AMPK might provide a new strategy for the management of hepatic encephalopathy. However, unselective drugs such as AICAR, which activate AMPK under normal as well as under stress conditions, are not suitable for clinical use. THC, the main active constituent of marijuana, has been repeatedly demonstrated to cause brain dysfunction and neurotoxicity (Mishima et al., 2001). This finding is in line with our observations disclosed herein below of its ability to stimulate AMPK. In addition, these studies have used high dosage of THC (1-15 mg/kg), quantities that we found necessary for AMPK activation under normal circumstances.
  • It is a finding of the present invention that the quantity of THC required to activate AMPK drops (from 10 mg/kg in healthy animals to 0.1 mg/kg in experimental hepatic encephalopathy animals) as shown herein below. Therefore, THC could be suitable as a selective agent that could function as a “stress specific drug” by activating AMPK only under pathological conditions.
  • The surprising fact that THC, which has about equal affinity for the CB1 and CB2 receptor, was effective in the treatment of induced hepatic encephalopathy in animal models, motivated us to investigate other compounds known to interact with receptors other than the endocannabinoid receptors. For example, capsaicin, suggested by Di Marzo et al (1998) to interact with the endocannabinoid system, acts on neural cells via vanilloid receptors subtype 1 (VR1, also known as transient receptor potential 1 TRPV1), a non-selective cation channel, which can be blocked by capsazepine. As shown herein below, capsaicin treatment of induced hepatic encephalopathy in animal models resulted in both improved hepatic and brain functions.
  • A second compound tested herein to treat the animals is cannabidiol, an active ingredient of Cannabis Sativa devoid of adverse effects related to the CB1 receptor owing to its CB1-independent mechanism of action. Cannabidiol is also a very potent anti-inflammatory agent. It is a finding of the present invention that cannabidiol improves impaired brain and liver function in experimental hepatic encephalopathy in animal models. Furthermore, two additional cannabinoids, 2-AG and HU-308 were shown herein to positively affect liver function in experimental hepatic encephalopathy in animal models.
  • The present invention thus provides a compound selected from D9-tetrahydrocannabinol (THC), cannabidiol and capsaicin for prevention, treatment, or both of hepatic encephalopathy.
  • In one preferred embodiment the compound is D9-tetrahydrocannabinol. In another preferred embodiment the compound is cannabidiol. In still another preferred embodiment the compound is capsaicin.
  • The compound may be formulated in any suitable form for administration, preferably in an oral, parenteral, sublingual or intranasal dosage form.
  • According to the present invention, D9-tetrahydrocannabinol, cannabidiol and capsaicin are intended for prevention and/or treatment of all subclasses of hepatic encephalopathy as described above, i.e. Type A, Type B or Type C, preferably type A or type C.
  • The present invention further provides a pharmaceutical composition for prevention, treatment, or both, of hepatic encephalopathy comprising a compound selected from D9-tetrahydrocannabinol, cannabidiol and capsaicin and a pharmaceutically acceptable carrier.
  • The present invention also concerns a method for prevention, treatment, or both, of hepatic encephalopathy comprising administering to a subject in need a therapeutically effective amount of a compound selected from D9-tetrahydrocannabinol (THC), cannabidiol and capsaicin.
  • In one aspect the present invention relates to a compound selected from capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 or cannabidiol for prevention, treatment, or both, of liver cirrhosis.
  • The invention further relates to a pharmaceutical composition comprising a compound, selected from capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 or cannabidiol for prevention, treatment, or both, of liver cirrhosis
  • In one preferred embodiment the compound is 2-arachidonoylglycerol. In another preferred embodiment the compound is HU-308. In still another preferred embodiment the compound is capsaicin. In yet another preferred embodiment the compound is cannabidiol.
  • The term “prevention of liver cirrhosis” refers herein to preventing or slowing the deterioration of any damage caused to the liver tissue, such as the accumulation of fibrotic scar tissue, by factors known to cause cirrhosis such as, but not limited to, alcoholic liver disease, chronic viral hepatitis type C, chronic viral hepatitis type B, chronic bile duct blockage, Wilson's disease, hemochromatosis, exposures to drug and toxins, autoimmune hepatitis, cystic fibrosis, alpha antitrypsin deficiency, obesity or schistosomiasis.
  • It is envisioned that prevention of the development of liver cirrhosis can be achieved by treating subjects in need, such as alcoholics, people infected with hepatitis C and obese people, at very early stages of their disease or condition, even before appearance of physical symptoms of liver cirrhosis.
  • The present invention also concerns a method for prevention, treatment, or both, of liver cirrhosis, comprising administering to a subject in need a therapeutically effective amount of a compound selected from capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 or cannabidiol.
  • The present invention further provides a compound selected from cannabidiol (CBD) or capsaicin for prevention, treatment, or both, of hepatic encephalopathy or liver cirrhosis.
  • The invention further provides a pharmaceutical composition for prevention, treatment, or both, of hepatic encephalopathy or liver cirrhosis, comprising cannabidiol or capsaicin and a pharmaceutically acceptable carrier.
  • The invention will now be illustrated by the following non-limiting examples.
  • EXAMPLES Materials and Methods
  • (i) Reagents. THC, SR141716A, SR144528 and CBD were provided by Prof. Raphael Mechoulam (Faculty of Medicine and Department of Pharmacology, Hebrew University of Jerusalem). Hepatotoxin thioacetamide (TAA) and capsaicin were obtained from Sigma-Aldrich (Rehovot, Israel). 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) was obtained from Toronto Research Chemicals (TRC). HU-308 was synthesized as described in Hanus et al. (1999).
  • (ii) Mice. Eight- to 10-week old female Sabra mice (29-32 g), obtained from the animal facility of the Hebrew University, Israel, were assigned at random to different groups of 10 mice per cage and were used in all experiments. All cages contained wood-chip bedding and were placed in a temperature-controlled room at 22° C., on a 12 h light/dark cycle (lights on at 07.00 a.m.). The mice had free access to water 24 h a day. The food provided was Purina chow, the animals were maintained in the animal facility (SPF unit) of the Hebrew University Hadassah Medical School, Jerusalem. CB-2 KO mice were provided by Prof. Zimmer, Institute of Molecular Psychiatry, University of Bonn, Germany.
  • Mice were sacrificed after treatment by decapitation between 10.00-12.00 a.m. Brains were rapidly removed and were dissected out and kept at −70° C.
  • (iii) Induction of Hepatic Failure
  • (iiia) Bile duct ligation. A midline incision was made under general anesthesia. The common bile duct was localized, doubly ligated, and cut between these two ligatures. In sham animals, a midline incision was performed, but with BDL.
  • (iiib) TAA. A single dose of 200 mg kg-1 of TAA was injected by the intraperitoneal route (i.p.). 24 hours after injection all animals (including control) were injected (s. c) with 0.5 ml solution of 0.45% NaCl, 5% dextrose and 0.2% KCl in order to prevent hypovolemia, hypokalemia and hypoglycemia. The mice were intermittently exposed to infrared light in order to prevent hypothermia. THC was administered i.p. either alone or with SR141716A on day 6 after TAA administration. Mice were sacrificed 1 h post treatment and analyzed for AMPK level. For the behavioral tests which started on day 6 after TAA administration, THC was administered i.p. during days 6-10. Neurological score, activity and cognitive function were analyzed during these days.
  • (iv) Immunoblot analysis. Total hippocampal protein was extracted using TriFast reagent (peqLab, Germany). Aliquots of the clarified lysates containing 30 mg protein were denatured in Laemmli sample buffer (6% SDS 30%, glycerol, 0.02% bromophenol blue, 200 mM Tris-HCl (pH 6.8), and 250 mM-mercaptoethanol, at 95° C. for 5 min. The samples were resolved by SDS—PAGE (10% acrylamide), and blotted onto nitrocellulose membrane. Non-specific binding in a Western blot analysis was prevented by immersing the membranes in blocking buffer (5% nonfat dry milk in Tris-buffer saline-Tween 20 (TBS-T)), for 2 h at room temperature. The membranes were then exposed to the indicated antibodies diluted 1:1000 for 1 h at room temperature. Anti-AMPK and phospho-AMPK antibodies were obtained from Cell Signaling. Anti-protein kinase B (AKT) was obtained from Upstate. Anti-actin was from Santa Cruz Biotechnology Inc. (Santa Cruz, Calif.). The blots were rinsed in TBS-T and then incubated with horseradish peroxidase-conjugated goat anti-mouse antibodies (1:10,000; Santa Cruz Biotechnology Inc., Santa Cruz, Calif.) for 1 h at room temperature. Antibody-antigen complexes were visualized by detecting enhanced chemiluminescence with X-ray film.
  • (v) RT-PCR analysis. Total hippocampal RNA was extracted using TriFast reagent according to the manufacturer's instructions and reverse transcribed. Primers specific for CB1 were GGAGAACATCCAGTGTGGGG [SEQ ID NO: 1] and CATTGGGGCTGTCTTTACGG [SEQ ID NO: 2], for CB2 GGGTCCTCTCAGCATTGATTT [SEQ ID NO: 3], and GTTAACAAGGCACAGCATGGAAC [SEQ ID NO: 4], and for actin CAG CTTCTTTGCAGCTCCTT [SEQ ID NO: 5] and TCACCCACATAGGAGTCCT [SEQ ID NO: 6]. All primers were synthesized by Danyel Biotech, Israel.
  • (vi) Catecholamine measurements. Catecholamines were measured as described previously (Avraham et al, 1996). The assay for dopamine was performed by High Pressure Liquid Chromatography (HPLC) separation and detection using HPLC-electrochemical detection (ECD). Values are presented as concentration (ng/g tissue).
  • (vii) Neurological function. Neurological function was assessed by a 10 point scale based on reflexes and task performance (Chen et al., 1996): exit from a circle 1 meter in diameter in less than 1 minute, seeking, walking a straight line, startle reflex, grasping reflex, righting reflex, placing reflex, corneal reflex, maintaining balance on a beam 3, 2 and 1 cm in width, climbing onto a square and a round pole. For each task failed or abnormal reflex reaction a score of 1 was assigned. Thus, a higher score indicates poorer neurological function. The neurological score was assessed one day after TAA induction (day 2). The mice were then divided between treatment groups so that each group had a similar baseline neurological score after TAA induction. The post-treatment neurological score was assessed one day after administration of the agonist or the antagonist or the vehicle (day 3).
  • (viii) Activity. The activity test was performed on day 4, since in the first 3 days after TAA injection almost no motor activity was observed. One of two methods was utilized: a) an activity apparatus, which consists of a cylindrical chamber (60 cm in diameter) with crossing infrared beams. Locomotor activity was recorded by a counter (attached to the apparatus), that counts the number of beam crossings made by the mice at one-minute intervals. Activity of two mice was measured simultaneously for a five-minute period. Two mice were tested together to lower stress to the minimum. Activity is presented as the mean number of beam crossings in 5 minutes.
  • Activity was also assessed in the open field (20×30 cm field divided into 12 squares of equal size) as described previously (Fride and Mechoulam, 1993). Two mice were observed simultaneously for 5 minutes. Locomotor activity was recorded by counting the number of crossings by the mice at one minute intervals. Results are presented as the mean number of crossings per minute.
  • (ix) Eight-arm maze. The animals were placed in an eight-arm maze, which is a scaled-down version of that developed for rats (Olton and Samuelson, 1976; Pick and Yanai, 1983). We used water deprivation achieved by limiting water consumption overnight and a reward of 50 μl of water presented at the end of each arm. The mice were tested until they made entries into all eight arms or until they completed 24 entries, whichever came first. Hence, the lower the score, the better the performance. Maze performance was calculated each day for five consecutive days. Results were presented as area under the curve (AUC) utilizing the formula: (day 2+day 3+day 4+day 5)−4*(day 1).
  • (x) Statistical analysis. Data are presented as means and standard deviations (SD) or standard errors (SEM). Results were evaluated by one-way ANOVA and 2-tailed t-test. Post-hoc testing was carried out using the Tukey-Kramer multiple comparisons procedure.
  • (xi) Liver function analysis. Ammonia, bilirubin, ALT, AST, GGT and glucose were analyzed using standard analytical methods in the Hadassah Hospital Biochemistry Department, Jerusalem, Israel.
  • Example 1 Experimental Hepatic Encephalopathy is Accompanied by Activation of AMPK by Cannabinoids
  • To consider their role in AMPK stimulation, we studied the effects of giving exogenous cannabinoids to activate AMPK. In the first step, control mice were administrated with 0.01 to 10 mg/kg THC and hippocampal AMPK phosphorylation was analyzed. THC treatment showed a biphasic effect (Sulcova et al., 1998). While low levels of THC (less than 0.1 mg/kg) reduced the level of activated AMPK, higher concentrations exhibited a dose dependent elevation in activated enzyme, reaching a significant activation of AMPK (FIG. 1). In the next step, the effect of THC was tested in TAA treated mice. In this instance THC also demonstrated a biphasic effect. However, an inactivating effect was already observed in 0.01 mg/kg and AMPK activation was achieved by 0.1 mg/kg. Elevation of the cerebral responsiveness to THC suggested that low doses of THC, which do not activate the AMPK in the healthy animals, could be used in the pathological state.
  • Example 2 THC Activates AMPK and Improves Impaired Brain Function in Experimental Hepatic Encephalopathy
  • Since treatment of 0.1 mg/kg THC augmented AMPK activation in a similar manner to AICAR treatment, we chose this dose to test THC's physiological effects on the experimental hepatic encephalopathy. TAA treated mice were administrated daily with 0.1 mg/kg THC for 5 days. Amplification of AMPK activation in response to THC administration was confirmed in the brains of the experimental animals at the end of the behavioral studies (FIG. 2A).
  • Next, we investigated the outcome of AMPK activation increase on brain function. Following the treatment, TAA-induced impaired cognitive function was improved significantly (FIG. 2B), poor activity performances were restored (FIG. 2C) and the reduced neurological score was improved (FIG. 2D). To reveal the mechanism by which THC could improve brain function, we studied the catecholaminergic response to THC treatment. Brain tissue in animals with experimental hepatic encephalopathy exhibited reduced dopamine concentrations while THC administration, similarly to AICAR administration, restored levels to normal (FIG. 2E). These results demonstrated the potential of THC to stimulate cerebral AMPK activity in treating hepatic encephalopathy.
  • Example 3 AICAR and THC Treatment do not Improve Markers of Hepatic Function
  • To investigate the possibility that the neural benefits of AICAR and THC might also result from peripheral effects (i.e. improvement of liver function) rather than cerebral function, we studied their effects on liver function. Animals treated with TAA exhibited hyperammonemia as a result of the liver dysfunction; AICAR and THC treatment had no effect on the ammonia level (FIG. 3A). Bilirubin levels and liver enzymes activity are the most commonly used laboratory markers of liver function. TAA treated mice demonstrated increased levels of bilirubin (FIG. 3B), alanine transaminase (ALT) (FIG. 3C), aspartate aminotransferase (AST) (FIG. 3D) and gamma-glutamyltransferase (GGT) (FIG. 3E). Neither AICAR nor THC ameliorated these markers indicating lack of direct action on liver recovery. Glucose analysis revealed a systemic hypoglycemia following TAA treatment (FIG. 3F) providing additional evidence for the metabolic energy impairment characterizing experimental hepatic encephalopathy.
  • Example 4 Capsaicin Improves Impaired Markers of Hepatic Function in Experimental Hepatic Encephalopathy
  • The surprising fact that THC, which has about equal affinity for the CB1 and CB2 receptor, was effective in the treatment of induced hepatic encephalopathy in animal models, motivated us to investigate other compounds known to interact with receptors other than the endocannabinoid receptors. For example, capsaicin, suggested by Di Marzo et al (1998) to interact with the endocannabinoid system, acts on neural cells via vanilloid receptors subtype 1 (VR1, also known as transient receptor potential 1 TRPV1), a non-selective cation channel, which can be blocked by capsazepine. Thus, TAA treated mice were administered different agonist/antagonists and capsaicin and their effect on hepatic function was assessed.
  • First, histopathological changes were observed after the treatment with TAA. FIGS. 4A-F depict varying degrees of necrosis which were semi-quantitated as histopathological indices 1-4.
  • Second, the effect on hepatic function of different agents administered to the TAA treated animals was tested. Comparison of the histopathological indices assessed for each group indicated that amelioration of TAA-induced apoptosis/necrosis reached statistical significance (P<0.05) only with capsaicin treatment. However, inflammation was reduced also in HU308 and SR141716A—treated animals when compared to TAA-treated group, whereas, SR144528 treatment resulted in non-significant changes with regard to the inflammatory process.
  • The regenerative capacity of the liver in all cannabinoid receptor agonists or antagonists—treated groups was higher when compared to animals to which only TAA was administered, except capsaicin treated animals which exhibited significantly less hepatic regeneration (Table 1).
  • TABLE 1
    Comparison of the frequency of cells showing apoptosis/necrosis,
    inflammation and regeneration in the liver.
    Groups
    comparison Apoptosis Inflammation Regeneration
    TAA vs normal Increase Increase Increase
    (p < 0.001*) (p < 0.001*) (p < 0.001*)
    TAA + capsaicin Decreased Decreased Decreased
    vs TAA (p < 0.005*) (p < 0.01*) (p < 0.001*)
    TAA + NS** Decreased Increased
    SR141716A vs (p < 0.001**) (p < 0.005*)
    TAA
    TAA + HU-308 NS* Decreased Increased
    vs TAA (p < 0.005*) (p < 0.001*)
    TAA + NS* NS* Increased
    SR144528 vs (p < 0.05*)
    TAA
    TAA + 2Ag vs NS* Decreased Increased
    TAA (p < 0.05*) (p < 0.05*)
    TAA + 2Ag + SR1 NS* Decreased Increased
    41716A vs TAA (p < 0.05*) (p < 0.05*)
    *= Fisher's exact test;
    **= Pearson's chi-square test
  • Example 5 CB1 Antagonist and CB2 Agonist Treatment Improve Markers of Hepatic Function
  • TAA treated mice demonstrated increased levels of alanine transaminase (ALT) (FIG. 5A) (see also FIG. 3C) and aspartate aminotransferase (AST) (FIG. 5B) (see also FIG. 3D); treatment of the TAA treated mice with 2-AG—a CB1 agonist, SR141716A—a CB1 antagonist, HU-308—a CB2 agonist, and SR144528—a CB2 antagonist, all significantly reduced ALT and AST levels. Moreover, 2-AG did not counteract the effect of SR141716A or SR144528, and HU-308 did not counteract the effect of SR144528. Thus, the results imply that the agonists/antagonists did not convey their effect specifically through the CB1 or CB2 receptors.
  • Example 6 CB2 Agonist, but not CB1 Antagonist Treatment Improves Markers of Hepatic Function in CB2-KO Mice
  • In the transgenic mice lacking the CB2 receptor, 2-AG but not SR141716A modestly but significantly reduced the ALT level and SR141716A blocked the effect of 2-AG (FIG. 6A), confirming that effect was achieved through the CB1 receptor.
  • The effect of the endocannabioid agonist and antagonist on the second hepatic function marker tested, AST, was inconsistent with the result obtained for ALT in that both the CB1 antagonist SR141716A and the CB1 agonist 2-AG were effective in reducing its level (FIG. 6B). SR141716A abolished the effect of 2-AG, or vice-versa.
  • Example 7 Capsaicin Treatment Improves Markers of Hepatic Function
  • The results disclosed above imply that the effect observed with the endocannabioid agonist and antagonist may have been affected through another receptor than the CB1 receptor. We therefore tested the effect of capsaicin, which as mentioned above, has been suggested to interact with the endocannabinoid system. FIGS. 7A-B show that indeed, capsaicin significantly reduces both the ALT and the AST level in TAA treated mice. The effect of capsaicin is specifically affected through the VR1 receptor as evidenced by the abolishment of the effect of capsaicin by the VR1 antagonist capsazepine.
  • Example 8 Endocannabioid Agonist and Antagonist and Capsaicin Treatment Reduce Astrogliosis in TAA Treated Mice
  • To assess whether endocannabioid agonist and antagonist and capsaicin treatment affect the important aspect of hepatic encephalopathy pathology—astrogliosis—TAA treated animals were treated with these compounds and hippocampus was stained for glial cells in naïve animals (FIG. 8A) and after treatment (FIGS. 8B-F).
  • It was found that hepatic encephalopathy induced intensive glial fibrillary acidic protein (GFAP, a marker for glial cells) staining intensity and increased process complexity, i.e. more processes and increased branching (FIG. 8B) as compared with naïve animals (FIG. 8 A). These changes were prevented following capsaicin—(FIG. 8C), CB1 antagonist (SR141716A)—(FIG. 8D) and CB2 agonist (HU-308)—(FIG. 8E) treatment, whereas CB2 antagonist (SR144528) administration (FIG. 8F) failed to prevent the hepatic encephalopathy induced changes and did not alter either the number or morphology of activated astroglia compared to untreated animals (FIG. 8B), as can be seen in Table 2, presenting the changes in quantitative terms.
  • TABLE 2
    Quantification of the changes in astrogliosis
    following TAA and treatments with various compounds.
    (two-sided Fisher's exact test)
    Comparison Direction of change
    TAA vs normal Increase (p < 0.001)
    TAA + capsaicin vs TAA Decrease (p < 0.001)
    TAA + SR141716A vs TAA Decrease (p < 0.001)
    TAA + HU-308 vs TAA Decrease (p < 0.001)
    TAA + SR144528 vs TAA NS
  • Example 9 Capsaicin Improves Impaired Brain Function in Experimental Hepatic Encephalopathy
  • Since treatment with capsaicin reduced astrogliosis in TAA treated mice, we were interested in assessing whether the treatment also had a positive effective on the impaired brain functions of hepatic encephalopathy mice. Following the treatment with capsaicin, TAA-induced impaired cognitive function was improved significantly (FIG. 9A), poor activity performances were restored (FIG. 9B) and the reduced neurological score was improved (FIG. 9C).
  • Example 10 2-AG Treatment Effect on Cognitive Impairment Secondary to Biliary Cirrhosis and Motor Impairments
  • As can be seen in FIGS. 10A-B, 2-AG effectively reversed cognitive impairments secondary to biliary cirrhosis in mice (FIG. 10A), but failed to reverse the motor impairments (FIG. 10B) which are also typical to this disorder.
  • Example 11 Cannabidiol Improves Impaired Brain and Liver Function in Experimental Hepatic Encephalopathy
  • We decided to treat the animals with cannabidiol, an active ingredient of Cannabis Sativa devoid of adverse effects related to the CB1 receptor owing to its CB1-independent mechanism of action. Cannabidiol is also a very potent anti-inflammatory agent.
  • Chronic liver failure was induced by bile duct ligation (BDL) in female Sabra mice. Sham-operated mice served as controls. BDL animals were divided randomly to control and treatment groups, which received, respectively, saline and 5 mg/kg cannabidiol i.p. daily for 3 weeks.
  • Two weeks post-surgery, the cognitive and the motor function of the mice were evaluated. Mice were decapitated 3 weeks post-surgery and their hippocampi were taken for analysis of IL-1b mRNA level by RT-PCR. The results clearly show that cognitive function (FIG. 11A) and motor activity (FIG. 11B) are impaired by BDL after 2 weeks and is restored by cannabidiol. Also, IL-1β mRNA level (normalized to ribosomal protein L19 mRNA levels; a commonly used invariant control gene) in the hippocampus is elevated following BDL and is restored by cannabidiol (FIGS. 12A-B).
  • Oxidative stress in liver tissue due to chronic liver failure induced by BDL was assessed by measuring malondialdehyde, a well accepted biomarker for oxidative stress. As can be seen in FIG. 13, oxidative stress is elevated following BDL and is restored by cannabidiol. As oxidative stress is commonly known to be involved in the development of cirrhosis (Ara et al., 2005) and treatment with cannabidiol reduces the oxidative stress, one can deduce that this treatment will prevent or slow down the development of cirrhosis.
  • In summary, after 2 weeks, bile duct ligation induced cognitive and motor deficits and increased oxidative stress in the liver, which were reversed by cannabidiol. In the hippocampus, which is responsible for learning and memory, there was an up-regulation of IL-1b mRNA following BDL, which was also reversed by cannabidiol, suggesting causal relationship between an inflammatory response in this region and impaired learning.
  • REFERENCES
    • Ara C, Kirimlioglu H, Karabulut A B, Coban S, Ay S, Harputluoglu M,
    • Kirimlioglu V, Yilmaz S. (2005) Protective effect of resveratrol against oxidative stress in cholestasis. J Surg Res. 127(2): 112-7
    • Avraham Y, Bonne O, Berry E M. Behavioral and neurochemical alterations caused by diet restriction—The effect of tyrosine administration in mice. Brain Research 1996; 732:133-144.
    • Avraham Y, Israeli E, Gabbay E, Okun A, Zolotarev O, Silberman I, Ganzburg V, Dagon Y, Magen I, Vorobia L, Pappo O, Mechoulam R, Ilan Y, Berry E M. Endocannabinoids affect neurological and cognitive function in thioacetamide-induced hepatic encephalopathy in mice. Neurobiol Dis. 2006 January; 21(1):237-45.
    • Baker D, Pryce G, Giovannoni G, Thompson A J. The therapeutic potential of cannabis. Review. Lancet Neurol 2003; 2:291-298.
    • Barbiroli B, Gaiani S, Lodi R, Totti S, Tonon C, Clementi V, Donati G, Bolondi L. Abnormal brain energy metabolism shown by in vivo phosphorus magnetic resonance spectroscopy in patients with chronic liver disease. Brain Res Bull 2002; 59:75-82.
    • Bergold P J, Sweatt J D, Winicov I, Weiss K R, Kandel E R, Schwartz J H. Protein synthesis during acquisition of long-term facilitation is needed for the persistent loss of regulatory subunits of the Aplysia cAMP-dependent protein kinase. Proc Natl Acad Sci USA. 1990; 87:3788-91.
    • Bernard A, Roger D, Luigi C, and Ramon L. Actinomycin D Blocks Formation of Memory of shock-avoidance in Goldfish. Science 1967; 158: 3808, 1600-1601
    • Berry E M, Mechoulam R. Tetrahydrocannabinol and endocannabinoids in feeding and appetite. Review. Pharmacol Ther 2002; 95:185-190.
    • Bezuglov V, Bobrov M, Gretskaya N, Gonchar A, Zinchenko G, Melck D, Bisogno T, Di Marzo V, Kuklev D, Rossi J C, Vidal J P, Durand T. Synthesis and biological evaluation of novel amides of polyunsaturated fatty acids with dopamine. Bioorg Med Chem Lett 2001; 1.1:447-449
    • Bisogno T, Melck D, Bobrov M Yu, Gretskaya N M, Bezuglov U V, De Petrocellis L, Di Marzo V. N-Acyl-dopamines: novel synthetic CB(1) cannabinoid-receptor ligands and inhibitors of anandamide inactivation with cannabimimetic activity in vitro and in vivo. Biochem J 2000; 351:817-824.
    • Cheer J F, Wassum K M, Heien M L, Phillips, P E, Wightman, R M. Cannabinoids enhance subsecond dopamine release in the nucleus accumbens of awake rats. J Neurosci 2004; 24:4393-4400.
    • Chen Y, Constantini S, Trembovler V, Weinstock M, Shohami E. An experimental model of closed head injury in mice: pathophysiology, histopathology and cognitive deficits. J Neurotrauma 1996; 13:557-568.
    • Costa-Mattioli M, Gobert D, Harding H, Herdy B, Azzi M, Bruno M, Bidinosti M, Ben Mamou C, Marcinkiewicz E, Yoshida M, Imataka H, Cuello A C, Seidah N, Sossin W, Lacaille J C, Ron D, Nader K, Sonenberg N. Translational control of hippocampal synaptic plasticity and memory by the eIF2alpha kinase GCN2. Nature. 2005; 436:1166-73.
    • Dagon Y, Avraham Y, Berry E M. AMPK activation regulates apoptosis, adipogenesis, and lipolysis by eIF2alpha in adipocytes. Biochem Biophys Res Commun. 2006; 340:43-7.
    • Dagon Y, Avraham Y, Magen I, Gertler A, Ben-Hur T, Berry E M. Nutritional status, cognition, and survival: a new role for leptin and AMP kinase. J Biol Chem 2005; 280:42142-42148.
    • Devane W A, Hanus L, Breuer A, Pertwee R G, Stevenson L A, Griffin G, Gibson D, Mandelbaum A, Etinger A, Mechoulam R. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992; 258:1946-1949.
    • Folbergrova J, Zhao Q, Katsura K, Siesjo B K. N-tert-butyl-alpha-phenylnitrone improves recovery of brain energy state in rats following transient focal ischemia. Proc Natl Acad Sci USA. 1995; 92:5057-5061.
    • Foretz M, Taleux N, Guigas B, Horman S, Beauloye C, Andreelli F, Bertrand L, Viollet B. (2006) Regulation of energy metabolism by AMPK: a novel therapeutic approach for the treatment of metabolic and cardiovascular diseases. Med Sci (Paris) 22(4):381-8.
    • Fride E, Mechoulam R. Pharmacological activity of the cannabinoid receptor agonist, anandamide, a brain constituent. Eur J Pharmacol 1993; 231:313-314.
    • Gaoni Y and Mechoulam R Isolation, structure and partial synthesis of an active constituent of hashish. J Am Chem Soc 1964; 86:1646-7
    • Gerber T, Schomerus H. Hepatic encephalopathy in liver cirrhosis: pathogenesis, diagnosis and management. Review. Drugs 2000; 60:1353-1370.
    • Gerdeman G L, Partridge J G, Lupica C R, Lovinger D M. It could be habit forming: drugs of abuse and striatal synaptic plasticity. Review. Trends Neurosci 2003; 26: 184-192.
    • Hanus, A. Breuer, S. Tchilibon, S. Shiloah, D. Goldenberg, M. Horowitz, R. G. Pertwee, R. A. Ross, R. Mechoulam, and E. Fride (1999) PNAS 96:14228-14233.
    • Hardie D G, Carling D. The AMP-activated protein kinase—fuel gauge of the mammalian cell? Review. Eur J Biochem 1997; 246:259-273.
    • Hardie D G. The AMP-activated protein kinase pathway—new players upstream and downstream. Review. J Cell Sci 2004; 117:5479-5487.
    • Hawley S A, Davison M, Woods A, Davies S P, Beri R K, Carling D, and Hardie D G. Characterization of the AMP-activated protein kinase kinase from rat liver and identification of threonine 172 as the major site at which it phosphorylates AMP-activated protein kinase. J Biol Chem 271: 27887-27879, 1996
    • Hoyer S. Abnormalities in brain glucose utilization and its impact on cellular and molecular mechanisms in sporadic dementia of Alzheimer type. Ann N Y Acad Sci 1993; 695:77-80.
    • Iversen L L. The Science of Marijuana (Oxford Univ. Press, New York, 2000), p. 36.
    • Julien B, Grenard P, Teixeira-Clerc F, Van Nhieu J T, Li L, Karsak M, Zimmer A, Mallat A, Lotersztajn S. Antifibrogenic role of the cannabinoid receptor CB2 in the liver. Gastroenterology. 2005; 3:128:742-55.
    • Kola B, Hubina E, Tucci S A, Kirkham T C, Garcia E A, Mitchell S E, Williams L M, Hawley S A, Hardie D G, Grossman A B, Korbonits M. Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. J Biol Chem. 2005; 280:25196-201.
    • Martin B R. Role of lipids and lipid signaling in the development of cannabinoid tolerance. Review. Life Sci 2005; 77:1543-1558.
    • Matsuda L A, Lolait S J, Brownstein M J, Young A C, Bonner T I. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990; 346:561-564.
    • Minokoshi Y, Alquier T, Furukawa N, Kim Y B, Lee A, Xue B, Mu J, Foufelle F, Ferre P, Birnabaum M J, Stuck B J, Kahn B B. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 2004; 428:569-574.
    • Mishima K, Egashira N, Hirosawa N, Fujii M, Matsumoto Y, Iwasaki K, Fujiwara M. Characteristics of learning and memory impairment induced by delta9-tetrahydrocannabinol in rats. Jpn J Pharmacol. 2001; 87:297-308.
    • Mizuno Y, Ikebe S, Hattori N, Nakagawa-Hattori Y, Mochizuki H, Tanaka M. Role of mitochondria in the etiology and pathogenesis of Parkinson's disease. Review. Biochim Biophys Acta 1995; 1271:265-274.
    • Molina-Holgado F, Pinteaux E, Heenan L, Moore J D, Rothwell N J, Gibson R M. Neuroprotective effects of the synthetic cannabinoid HU-210 in primary cortical neurons are mediated by phosphatidylinositol 3-kinase/AKT signaling. Mol Cell Neurosci. 2005; 28:189-94.
    • Olton D S, Samuelson R J. Remembrance of places passed: Spatial memory in rats. J Exp Psychol Anim Behav Process 1976; 2:97-116.
    • Ott P, Clemmesen O, Larsen F S. Cerebral metabolic disturbances in the brain during acute liver failure: from hyperammonemia to energy failure and proteolysis. Review. Neurochem Int 2005; 47:13-18.
    • Pagotto U, Vicennati V, Pasquali R. The endocannabinoid system and the treatment of obesity. Ann Med. 2005; 37:270-5. Review.
    • Pick C G, Yanai J. Eight arm maze for mice. Int J Neurosci 1983; 21:63-66.
    • Ross B D, et al. 31P spectroscopic imaging shows energy deficit of thalamus in chronic hepatic encephalopathy. In: Book of Abstracts, Annual Meeting of the Society of Magnetic Resonance in Medicine, Amsterdam; 1989: 465.
    • Sulcova E, Mechoulam R, Fride E. Biphasic effects of anandamide. Pharmacol Biochem Behav 1998; 59:347-353.
    • Teixeira-Clerc F, Julien B, Grenard P, Tran Van Nhieu J, Deveaux V, Li L, Serriere-Lanneau V, Ledent C, Mallat A, Lotersztajn S. CB1 cannabinoid receptor antagonism: a new strategy for the treatment of liver fibrosis. Nat Med. 2006; 6:12:671-6.
    • Xiong Y, Peterson P L, Lee C P. Alterations in cerebral energy metabolism induced by traumatic brain injury. Review. Neurol Res 2001; 23:129-138.
    • Zimmermann C, Ferenci P, Pifl C, Yurdaydin C, Ebner J, Lassmann H, Roth E, Hortangl H. Hepatic encephalopathy in thioacetamide-induced acute liver failure in rats: characterization of an improved model and study of amino acid-ergic neurotransmission. Hepatology 1989; 9:594-601.

Claims (11)

1. A method for prevention, treatment, or both, of hepatic encephalopathy, comprising administering to a subject in need a therapeutically effective amount of a compound selected from the group consisting of D9-tetrahydrocannabinol, cannabidiol and capsaicin.
2. The method according to claim 1, wherein said compound is D9-tetrahydrocannabinol.
3. The method according to claim 1, wherein said compound is administered orally, parenterally, sublingually, or by inhalation.
4. The method according to claim 1, wherein said hepatic encephalopathy is of Type A or Type C.
5. A method for prevention, treatment, or both, of liver cirrhosis, comprising administering to a subject in need a therapeutically effective amount of a compound selected from the group consisting of capsaicin, 2-arachidonoylglycerol (2-AG), HU-308 and cannabidiol.
6. The method according to claim 5, wherein said compound capsaicin.
7. The method according to claim 5, wherein said compound is administered orally, parenterally, sublingually, or by inhalation.
8. A method according to claim 1, wherein said compound is capsaicin.
9. A method for prevention, treatment, or both, of hepatic encephalopathy, comprising administering to a subject in need a therapeutically effective amount of D9-tetrahydrocannabinol.
10. A method for prevention, treatment, or both, of hepatic encephalopathy, comprising administering to a subject in need a therapeutically effective amount of capsaicin.
11. A method for prevention, treatment, or both, of liver cirrhosis comprising administering to a subject in need a therapeutically effective amount of capsaicin.
US12/429,832 2006-10-25 2009-04-24 Treatment of hepatic encephalopathy and liver cirrhosis Abandoned US20100022631A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/429,832 US20100022631A1 (en) 2006-10-25 2009-04-24 Treatment of hepatic encephalopathy and liver cirrhosis
US13/162,160 US10166202B2 (en) 2006-10-25 2011-06-16 Treatment of hepatic encephalopathy and liver cirrhosis

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US85407306P 2006-10-25 2006-10-25
US92944407P 2007-06-27 2007-06-27
US92944307P 2007-06-27 2007-06-27
PCT/IL2007/001300 WO2008050344A2 (en) 2006-10-25 2007-10-25 Treatment of hepatic encephalopathy and liver cirrhosis
US12/429,832 US20100022631A1 (en) 2006-10-25 2009-04-24 Treatment of hepatic encephalopathy and liver cirrhosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2007/001300 Continuation-In-Part WO2008050344A2 (en) 2006-10-25 2007-10-25 Treatment of hepatic encephalopathy and liver cirrhosis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/162,160 Division US10166202B2 (en) 2006-10-25 2011-06-16 Treatment of hepatic encephalopathy and liver cirrhosis

Publications (1)

Publication Number Publication Date
US20100022631A1 true US20100022631A1 (en) 2010-01-28

Family

ID=39125279

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/429,832 Abandoned US20100022631A1 (en) 2006-10-25 2009-04-24 Treatment of hepatic encephalopathy and liver cirrhosis
US13/162,160 Expired - Fee Related US10166202B2 (en) 2006-10-25 2011-06-16 Treatment of hepatic encephalopathy and liver cirrhosis

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/162,160 Expired - Fee Related US10166202B2 (en) 2006-10-25 2011-06-16 Treatment of hepatic encephalopathy and liver cirrhosis

Country Status (3)

Country Link
US (2) US20100022631A1 (en)
EP (1) EP2077828A2 (en)
WO (1) WO2008050344A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012144938A2 (en) 2011-03-16 2012-10-26 Общество С Ограниченной Ответственностью "Биотехнологии Пущино" Composition of protectors of acute and chronic hepatic encephalopathy and method for treating acute and chronic hepatic encephalopathy
WO2015106108A3 (en) * 2014-01-13 2015-11-12 Kannalife, Inc. Novel functionalized 1,3-benzene diols and their method of use for the treatment of hepatic encephalopathy
US9265458B2 (en) 2012-12-04 2016-02-23 Sync-Think, Inc. Application of smooth pursuit cognitive testing paradigms to clinical drug development
US9380976B2 (en) 2013-03-11 2016-07-05 Sync-Think, Inc. Optical neuroinformatics
US10610512B2 (en) 2014-06-26 2020-04-07 Island Breeze Systems Ca, Llc MDI related products and methods of use

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010012506A1 (en) * 2008-07-31 2010-02-04 Bionorica Research Gmbh Cannabinoids for use in treating or preventing cognitive impairment and dementia
EP2346519B1 (en) 2008-10-02 2015-12-02 George Zabrecky Methods and formulations for treating chronic liver disease
EP2456428A1 (en) * 2009-07-23 2012-05-30 INSERM - Institut National de la Santé et de la Recherche Médicale Selective cb2 receptor agonists for use in the prevention or treatment of alcoholic liver disease
CA3077624A1 (en) 2016-10-01 2018-04-05 James Smeeding Pharmaceutical compositions comprising a statin and a cannabinoid and uses thereof
WO2019121764A1 (en) * 2017-12-21 2019-06-27 Technische Universität Dresden Medicament for the treatment of liver fibrosis and cirrhosis of liver
JP2023551423A (en) * 2020-11-23 2023-12-08 ソシエテ・デ・プロデュイ・ネスレ・エス・アー Substituted resorcinol compounds, compositions, methods and uses thereof as AMPK activators

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0005789D0 (en) * 2000-03-11 2000-05-03 Knoll Ag Therapeutic agents
AU2001243369A1 (en) * 2000-03-14 2001-09-24 Massachusetts Institute Of Technology Composition and method to treat weight gain and obesity attributable to psychotropic drugs
US7320805B2 (en) 2003-10-01 2008-01-22 Institut National De La Sante Et De La Recherche Medicale CB2 receptors blocks accumulation of human hepatic myofibroblasts: a novel artifibrogenic pathway in the liver

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012144938A2 (en) 2011-03-16 2012-10-26 Общество С Ограниченной Ответственностью "Биотехнологии Пущино" Composition of protectors of acute and chronic hepatic encephalopathy and method for treating acute and chronic hepatic encephalopathy
US9265458B2 (en) 2012-12-04 2016-02-23 Sync-Think, Inc. Application of smooth pursuit cognitive testing paradigms to clinical drug development
US9380976B2 (en) 2013-03-11 2016-07-05 Sync-Think, Inc. Optical neuroinformatics
WO2015106108A3 (en) * 2014-01-13 2015-11-12 Kannalife, Inc. Novel functionalized 1,3-benzene diols and their method of use for the treatment of hepatic encephalopathy
CN106456573A (en) * 2014-01-13 2017-02-22 康纳生物技术有限公司 Novel functionalized 1,3-benzene diols for the treatment of hepatic encephalopathy and their method of use
JP2017507119A (en) * 2014-01-13 2017-03-16 カンナライフ・サイエンシーズ・インコーポレイテッドKannalife Sciences, Inc. Novel functional 1,3-benzenediols and methods for their use to treat hepatic encephalopathy
US9611213B2 (en) 2014-01-13 2017-04-04 Kannalife Sciences, Inc. Functionalized 1,3-benzene diols and their method of use for the treatment of hepatic encephalopathy
US10004722B2 (en) 2014-01-13 2018-06-26 Kannalife Sciences, Inc. Method for treating hepatic encephalopathy or a disease associated with free radical mediate stress and oxidative stress with novel functionalized 1,3-benzene diols
AU2015204609B2 (en) * 2014-01-13 2019-11-07 Kannalife Sciences, Inc. Novel functionalized 1,3-benzene diols and their method of use for the treatment of hepatic encephalopathy
US10610512B2 (en) 2014-06-26 2020-04-07 Island Breeze Systems Ca, Llc MDI related products and methods of use

Also Published As

Publication number Publication date
US10166202B2 (en) 2019-01-01
EP2077828A2 (en) 2009-07-15
WO2008050344A3 (en) 2008-08-28
US20110251290A1 (en) 2011-10-13
WO2008050344A2 (en) 2008-05-02

Similar Documents

Publication Publication Date Title
US10166202B2 (en) Treatment of hepatic encephalopathy and liver cirrhosis
Liu et al. Icariin exerts an antidepressant effect in an unpredictable chronic mild stress model of depression in rats and is associated with the regulation of hippocampal neuroinflammation
Feng et al. Curcumin prevents high-fat diet-induced hepatic steatosis in ApoE−/− mice by improving intestinal barrier function and reducing endotoxin and liver TLR4/NF-κB inflammation
Arora et al. Curcumin ameliorates reserpine-induced pain–depression dyad: Behavioural, biochemical, neurochemical and molecular evidences
Gibney et al. Inhibition of stress-induced hepatic tryptophan 2, 3-dioxygenase exhibits antidepressant activity in an animal model of depressive behaviour
Tiwari et al. Resveratrol abrogates alcohol-induced cognitive deficits by attenuating oxidative–nitrosative stress and inflammatory cascade in the adult rat brain
Rossetti et al. Oxidation-reduction mechanisms in psychiatric disorders: A novel target for pharmacological intervention
Ma et al. Quercitrin offers protection against brain injury in mice by inhibiting oxidative stress and inflammation
Wu et al. Neferine alleviates memory and cognitive dysfunction in diabetic mice through modulation of the NLRP3 inflammasome pathway and alleviation of endoplasmic-reticulum stress
Cippitelli et al. Protection against alcohol-induced neuronal and cognitive damage by the PPARγ receptor agonist pioglitazone
Xue et al. Melatonin mediates protective effects against kainic acid-induced neuronal death through safeguarding ER stress and mitochondrial disturbance
Droege et al. Aberrant insulin receptor signaling and amino acid homeostasis as a major cause of oxidative stress in aging
Uniyal et al. Co-treatment of piracetam with risperidone rescued extinction deficits in experimental paradigms of post-traumatic stress disorder by restoring the physiological alterations in cortex and hippocampus
Cline et al. Lasting downregulation of the lipid peroxidation enzymes in the prefrontal cortex of mice susceptible to stress-induced anhedonia
Kim et al. Theanine is a candidate amino acid for pharmacological stabilization of mast cells
Cheng et al. Protocatechuic acid-mediated DJ-1/PARK7 activation followed by PI3K/mTOR signaling pathway activation as a novel mechanism for protection against ketoprofen-induced oxidative damage in the gastrointestinal mucosa
Su et al. Formaldehyde as a trigger for protein aggregation and potential target for mitigation of age-related, progressive cognitive impairment
Zhang et al. Protective effects of kaempferol on D-ribose-induced mesangial cell injury
KR20130098347A (en) Methods of treating alcohol intoxication, alcohol use disorders and alcohol abuse which comprise the administration of dihydromyricetin
Kumar et al. Effect of curcumin on intracerebroventricular colchicine-induced cognitive impairment and oxidative stress in rats
Park et al. (S) YS-51, a novel isoquinoline alkaloid, attenuates obesity-associated non-alcoholic fatty liver disease in mice by suppressing lipogenesis, inflammation and coagulation
Cheng et al. Minocycline, a classic antibiotic, exerts psychotropic effects by normalizing microglial neuroinflammation-evoked tryptophan-kynurenine pathway dysregulation in chronically stressed male mice
Lo et al. Andrographolide inhibits IL-1β release in bone marrow-derived macrophages and monocyte infiltration in mouse knee joints induced by monosodium urate
Zhao et al. Malvidin alleviates mitochondrial dysfunction and ROS accumulation through activating AMPK-α/UCP2 axis, thereby resisting inflammation and apoptosis in SAE mice
Wang et al. Cannabidiol alleviates the damage to dopaminergic neurons in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced Parkinson’s disease mice via regulating neuronal apoptosis and neuroinflammation

Legal Events

Date Code Title Description
AS Assignment

Owner name: ARISTOTLE UNIVERSITY OF THESSALONIKI, GREECE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GRIGORIADIS, NICHOLAS;POUTACHIDIS, THEOFILOS;REEL/FRAME:023339/0225

Effective date: 20090801

Owner name: YISSUM RESEARCH DEVELOPMENT COMPANY OF THE HEBREW

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BERRY, ELLIOT;AVRAHAM, YOSEFA;MECHOULAM, RAPHAEL;AND OTHERS;REEL/FRAME:023339/0193

Effective date: 20090504

Owner name: HADASIT MEDICAL RESEARCH SERVICES & DEVELOPMENT LT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ILAN, YARON;REEL/FRAME:023339/0258

Effective date: 20090510

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: YISSUM RESEARCH DEVELOPMENT COMPANY OF THE HEBREW UNIVERSITY OF JERUSALEM LTD, ISRAEL

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE THE ASSIGNEE'S NAME PREVIOUSLY RECORDED AT REEL: 023339 FRAME: 0193. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNORS:BERRY, ELLIOT;AVRAHAM, YOSEFA;MECHOULAM, RAPHAEL;AND OTHERS;REEL/FRAME:060619/0450

Effective date: 20090504