GB2464813A - Treatments for neurodegenerative disorders - Google Patents

Treatments for neurodegenerative disorders Download PDF

Info

Publication number
GB2464813A
GB2464813A GB0918626A GB0918626A GB2464813A GB 2464813 A GB2464813 A GB 2464813A GB 0918626 A GB0918626 A GB 0918626A GB 0918626 A GB0918626 A GB 0918626A GB 2464813 A GB2464813 A GB 2464813A
Authority
GB
United Kingdom
Prior art keywords
none
oxidative stress
disease
nrf2
compounds
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.)
Withdrawn
Application number
GB0918626A
Other versions
GB0918626D0 (en
Inventor
Pamela Shaw
Richard Mead
Adrian Higginbottom
Sian Barber
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.)
University of Sheffield
Original Assignee
University of Sheffield
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 University of Sheffield filed Critical University of Sheffield
Priority to GB0918626A priority Critical patent/GB2464813A/en
Publication of GB0918626D0 publication Critical patent/GB0918626D0/en
Publication of GB2464813A publication Critical patent/GB2464813A/en
Withdrawn legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B35/00ICT specially adapted for in silico combinatorial libraries of nucleic acids, proteins or peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • 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
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/19Acanthaceae (Acanthus family)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6897Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids involving reporter genes operably linked to promoters
    • C40B30/02
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/06Methods of screening libraries by measuring effects on living organisms, tissues or cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5041Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects involving analysis of members of signalling pathways
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5058Neurological cells
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/60In silico combinatorial chemistry

Abstract

The present invention relates to the use of andrographolide or [S]+ apomorphine, which are Nrf2-ARE pathway activators, for the treatment of motor neurone disease. Andrographolide or [S]+ apomorphine are also suitable for the treatment of other neurodegenerative diseases known to be mediated by oxidative stress such as amyotrophic lateral sclerosis (ALS), primary lateral sclerosis (PLS), motor neurone disease, Huntington's disease, Alzheimer's disease or Parkinson's disease. These compounds may also be used to treat age-related macular degeneration, photogenic oxidative stress and skin ageing and radiation-induced cell damage, and they may be used for the preservation of organs for transplant and the stabilisation of cell cultures. The agents were identified by screening a library of agents for their suitability for the treatment of diseases mediated by oxidative stress, comprising exposing non-neuronal and neuronal cells (eg astrocytes) to the agents and identifying candidates that can activate the Nrf2-ARE pathway, and ascertaining physical and chemical parameters (eg the ability to cross the blood-brain barrier) by in silico methods.

Description

I
THERAPEUTICS FOR NEUROLOGICAL DISORDERS
The present invention relates to therapeutic agents for the treatment of neurological disorders known to be mediated by oxidative stress and in particular for the treatment of motor neurone disease and amyotrophic lateral sclerosis. The invention includes inter ella products for the treatment of neurological disorders.
BACKGROUND
Oxidative stress refers to the cytopathologic consequences of a mismatch between the production of free-radicals and the ability of the cell to defend against them. Growing data from experimental models and human brain studies suggest that oxidative stress may play an important role in neuronal degenerative diseases. Oxidative stress has been implicated in both normal aging and in various neurodegenerative disorders such as Parkinson's disease, Alzheimer's disease and amyotrophic lateral sclerosis and may be a common mechanism underlying various forms of cell death including necrosis, apoptosis, and excitotoxicity.
Motor neurone disease (MND) is more commonly called Amyotrophic lateral sclerosis (ALS) in the USA and is a progressive, fatal, neurodegenerative disease characterised by the loss of motor neurones in the motor cortex, brain stem and spinal cord, this leads to weakness and atrophy. ALS typically leads to death within 2-3 years of diagnosis.
ALS occurs in both sporadic (90% of all cases) and familial forms (10% of all cases). In 20% of familial ALS, mutations have been found in the copper, zinc superoxide dismutase gene (SOD1). The genes involved in the sporadic cases and in the remaining 80% of familial cases have yet to be identified. Currently there is no treatment which prevents or reverses the course of the disorder. Available treatments (such as riluzole and antioxidants) can at best only extend survival to a modest degree.
The mechanisms underlying the disease are not fully understood. However, the identification of SODI mutations as causative in a proportion of familial cases of ALS has allowed the generation of both cellular and mouse models of the disease which have greatly enhanced the understanding of disease mechanisms. Evidence from these models, as well as from patients, has provided very good evidence for a role of oxidative stress in disease pathogenesis. Oxidative stress has significant crosstalk to other potential mechanisms of neuronal injury, such as mitochondrial dysfunction, excitotoxicity, protein aggregation, cytoskeletal dysfunction and glial cell activation. It can feed into these mechanisms or is enhanced, in turn, by them. This central role in pathogenesis is reflected in a meta-analysis of studies in the most commonly used mouse model of ALS (transgenic mice expressing the G93A mutant form of human SODI) where therapies targeting oxidative stress have been highlighted as demonstrating greatest promise in slowing disease progression.
Despite this central role, targeting oxidative stress in ALS has failed to translate into clinical benefit for patients, which may in part be due to the lack of sufficiently potent anti-oxidants able to access the CNS. A novel approach to limiting oxidative stress in neurodegenerative disease is to promote activation of the transcription factor, NFE2-related factor 2 (Nrf2). Nrf2 drives expression of a battery of Phase II detoxification and anti-oxidant enzymes via its interaction with the antioxidant response element (ARE).
When activated, this programmed cell life' response is neuroprotective and conversely, attenuation of this pathway can enhance neuronal sensitivity to a range of neurotoxic challenges. Dysregulation of this pathway has been observed in ALS cellular models and confirmed in human tissue. Nrf2 itself and multiple target genes were repressed in a motor neuronal cell line expressing mutant (G93A) human SODi. Further, a target of this pathway which plays an important role in mitochondrial anti-oxidant defence, peroxiredoxin3 (PRDX3), was downregulated in both this cellular model and human tissue from familial and sporadic ALS. G93A SODI transgenic mice, when crossed with an ARE reporter mouse, showed activation of the Nrf2-ARE pathway in muscle from 30 days of age, with marginal activation in spinal cord at 90 days and more robust activation at the 110 day time-point. At 90 days of age, mice have already begun to show muscle weakness and motor neuron loss and at 110 days they show significant motor neuron pathology. This suggests that activation of the Nr12-ARE pathway in this murine model may be qualitatively insufficient or too late to protect motor neurons from significant damage. Taken in combination with other reports, this may reflect a defect in the capacity of cells expressing mutant 5001 to activate this pathway.
The Nrf2-ARE pathway is an attractive therapeutic target in ALS because it is well defined, amenable to activation by small molecules and activation of cellular defences may confer a more lasting protection against oxidative stress than, for example a direct scavenging approach. It is known from the prior art that a flavonoid derived from green tea catechin -(-) epigallocatechin-3-gallate (ECCO) shows neuroprotective effects in a motor neuronal cell line expressing mutant (G93A) human SOD1. This cell line was partially protected from H202 induced cell death at concentrations of ECCO greater than 2OpM. This compound has also been tested in the G93A mouse model of ALS at a range of doses, once a day, orally from 60 days of age. At higher doses a significant extension in survival was seen with an increase in mean survival. This therapeutic effect is found in spite of the fact that EGCG is itself pro-oxidant, narrowing its therapeutic window, and highly polar, making it unlikely to cross the blood brain barrier in significant concentrations.
There is a need for therapeutic agents that are effective in the treatment of diseases known to be mediated by oxidative stress and especially diseases like motor neurone disease, ALS, Parkinson's and other neurodegenerative diseases.
There is a need for therapeutic agents for the treatment of neurodegenerative diseases that possess minimal toxicity and have the ability to cross the blood-brain barrier and so can penetrate the CNS.
BRIEF SUMMARY OF THE DISCLOSURE
According to a first aspect of the invention there is provided a therapeutic agent for the treatment of motor neurone disease, the therapeutic agent being a Nrf2-ARE pathway activator selected from the group comprising andrographolide and S [+] apomorphine Motor Neurone Disease (MND) is an all embracing term used to cover a number of illnesses of the motor neurone. Amyotrophic Lateral Sclerosis (ALS), Progressive Muscular Atrophy (PMA), Progressive Bulbar Palsy (PBP), Primary Lateral Sclerosis (PLS) are all subtypes. MND is the generic term used more in Europe whilst ALS is sometimes used more generically in the USA. It will be appreciated by the skilled person that references to motor neurone disease (MND) also extend to references to ALS, PMA, PBP and PLS and these named disease states may be used interchangeably.
Throughout the description and claims of this specification, the words "comprise" and "contain" and variations of the words, for example "comprising" and "comprises", means "including but not limited to", and is not intended to (and does not) exclude other moieties, additives, components, integers or steps.
Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise.
Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith.
The present invention has demonstrated through a cascade of screens and tests that andrographolide and S [+] apomorphine are potent and drug-like' Nrf2-ARE activators which also have the capacity to penetrate the CNS.
The compounds of the present invention may be used prophylactically or therapeutically either on their own or as part of a treatment regimen.
It will be appreciated that the term "treatment" and "treating" as used herein means the management and care of a patient for the purpose of combating a condition, such as a disease or a disorder. The term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound to alleviate the symptoms or complications, to delay the progression of the disease, disorder or condition, to alleviate or relieve the symptoms and complications, andfor to cure or eliminate the disease, disorder or condition as well as to prevent the condition, wherein prevention is to be understood as the management and care of a patient for the purpose of combating the disease, condition, or disorder and includes the administration of the active compounds to prevent the onset of the symptoms or complications. The patient to be treated is preferably a mammal, in particular a human being, but it may also include animals, such as dogs, cats, cows, sheep and pigs.
According to a yet further aspect of the invention there is provided use of a Nrf2-ARE pathway activator selected from the group comprising andrographolide and S [+] apomorphine for the manufacture of a medicament for the treatment of motor neurone disease.
Apomorphine is a dopamine agonist typically administered subcutaneously as intermittent injections or in a continuous infusion. It is useful in managing advanced Parkinson's disease, and provides an alternative to neurosurgical procedures. There has been no previous clinical indication that [SI + enantiomer of the compound may be useful in treating ALS or other disease states. Results have shown that in particular the S enantiomer which does not have dopamine agonist activity has the correct criteria for being an ALS therapeutic as defined by the methods of the present invention and as described hereinbefore, The present invention therefore recognises new therapeutic effects of the [5] + enantiomer of apomorphine.
The chemical structure of apornorphine is given below and it will be appreciated that any variants and substitutions of the [51+ enantiomer is encompassed within the compounds of the present invention. In addition any derivatives or salts of the [S]+ enantiomer are included within the scope of the present invention and the contents of PCT1U503108448 are incorporated herein by reference.
HON
,H HCL
N
Andrographolide is a diterpenoid lactone of the plant Andrographis paniculata, known to possess anti-tumour activity for certain specific cancers such as breast cancer and to have an anti-inflammatory effect. There has been no previous clinical indication that the compound may be useful in treating diseases associated with oxidative stress for example and without limitation ALS. The present invention therefore recognises new therapeutic effects of andrographolide.
The chemical structure of andrographolide is given below and it will be appreciated that any variations and substitutions are encompassed within the compounds of the present invention. For example andrographolide and its derivatives may be of the formula where R.sub.1, R.sub.2 and R.sub.3 can independently represent hydrogen, acyl, phenyl, mono-or polyphosphate, mono-or polysulfate, glycosyl, cyclic or acyclic alkyl, alkenyl or alkynyl, wherein said phosphate or sulfate derivatives may be in the form of free acids or as salts.
Preferably, the compounds of the present invention may be formulated into pharmaceutical forms suitable for administration to a patient in need of treatment.
The pharmaceutical compositions can be in any form suitable for oral, parenteral, topical, intranasal, ophthalmic, otic, rectal or transdermal administration. Where the compositions are intended for parenteral administration, they can be formulated for intravenous, intramuscular, intraperitoneal, subcutaneous administration or for direct delivery into a target organ or tissue by injection, infusion or other means of delivery.
The delivery can be by bolus injection, short term infusion or longer term infusion and can be via passive delivery or through the utilisation of a suitable infusion pump.
Accordingly the compounds of the present invention may further include pharmaceutical ingredients or excipients.
"Pharmaceutical ingredient" or "excipient" means a pharmacologically inactive pharmaceutically acceptable compound added to the compositions of the inventiorn The ingredient or excipient does not have any pharmacological properties.
According to a further aspect of the invention there is provided a therapeutic agent for the treatment of a neurodegenerative condition that occurs as a result of oxidative stress, the therapeutic agent being a Nrf2-ARE pathway activator selected from the group comprising andrographolide and S [+1 apomorphine.
Preferably, the neurodegenerative condition know to be mediated by oxidative stress is selected from the group comprising motor neurone disease, amyotrophic lateral sclerosis (ALS) and primary lateral sclerosis (PLS), Huntington's disease, age related macular degeneration, preservation of organs for transplant/surgical procedures, stabilisation of cell cultures, photogenic oxidative stress and skin ageing and the treatment of radiation-induced cell damage.
According to a yet further aspect of the invention there is provided use of andrographolide for the treatment of a disease or condition that occurs as a result of oxidative stress, the disease or condition being selected from the group comprising motor neurone disease, amyotrophic lateral sclerosis (ALS), primary lateral sclerosis (PLS), Huntington's disease, Alzheimer's disease, Parkinson's disease, age related macular degeneration, preservation of organs for transplant/surgical procedures, stabilisation of cell cultures, photogenic oxidative stress and skin ageing and the treatment of radiation-induced cell damage.
According to a yet further aspect of the invention there is provided a method of treating an individual suffering from a neurodegenerative disease that occurs as a result of oxidative stress comprising administering a therapeutically effective amount of a Nrf2-ARE pathway activator selected from the group comprising andrographolide and S [1 apomorphine According to a yet further aspect of the invention there is provided an in vitro method of screening a library of candidate therapeutic agents for their suitability to treat diseases know to be mediated by oxidative stress, the method comprising: (i) exposing control or normal cells of non-neuronal origin to the candidate therapeutic and identifying candidate agents that possess an ability to activate the Nrf2-ARE pathway; (ii) exposing cells of neuronal origin with candidate agents having a positive effect in step (i) and assessing their ability to activate the Nr12-ARE pathway; (iii) assessing the ability of the candidate agent to protect cells of neuronal origin against an oxidative stress insult; and (iv) performing a series of in silico tests to ascertain physical and chemical parameters, wherein a candidate therapeutic having positive results in step (i-ui) and having suitable physico-chemical properties is likely to be suitable for the treatment of diseases know to be mediated by oxidative stress.
The present invention provides a convenient cascade of tests as a screening method for identifying more potent and drug-like' Nr12-ARE activators which also have the capacity to penetrate the CNS. The methods of the present invention provide a robust screening cascade to select a small number of promising molecules for further in vivo testing.
These "hit" molecules are tested initially for their capacity to activate the Nrf2-ARE pathway in cell lines derived from normal non-human animals and cell lines of human origin and subsequently used as "tool" molecules to determine whether the pathway could be activated in neuronal cells derived by primary culture from the CNS of non-human animals.
Preferably, the non-human animal from which cells are derived are selected from the group comprising monkeys, dogs, cats, rabbits, rats and mice. Rodent species are preferred, and the mouse is the most preferred species.
Preferably, in one embodiment of the invention the cells are stably transfected with an ARE reporter construct, optionally the reporter is a fluorescent agent such as and without limitation GFP or the like.
Preferably, cells are assayed for increased expression of NrI2 regulated genes. Suitable methods for detecting activation are described herein after.
Preferably, the cells of neuronal origin are CNS cells and especially are astrocytes. In one embodiment of the invention they are derived from transgenic non-human animals expressing the 093A mutant form of human SOD1.
Preferably, the oxidative stress test of step (iii) comprises withdrawal of serum and measurement of oxidative stress optionally with dichloroflourescein or derivatives or incubation with mitochondrial toxins (menadione) and measurement of cell survival.
However, it will be appreciated that other stress test can be employed in the methods of the present invention.
Preferably, the suitable physical and chemical parameters of step (iv) are a cLogP <5, molecular mass <500, <5 hydrogen bond donors (OH+NH count) and <10 hydrogen bond acceptors (0 plus N atoms). The suitable physical and chemical parameters are typically referred to as Lipinski's rule of 5. In addition AlogP less than 4 but greater than 1, and a molecular Polar Surface Areasbelow 100 (ideally 80) are optimal for passive diffusion across the blood-brain barrier (BBB) however further preferred characteristics may also be applied to best select candidate therapeutic agents.
Any features ascribed to a particular aspect of the invention are intended to apply to each and every aspect mutatis mutandis.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows a schematic representation of the NRF2-ARE assay used in the present invention.
Figure 2 shows the results of assay validation. Figure Ia shows concentration response curves for tBHQ (open squares) and EGCG (closed squares) in the CHO-4xARE-TK cell line. Both molecules have a narrow window of ARE activation peaking at I OisM and 1 00tM respectively. Figure lb shows results of Z' score determination. The average +1-SD for vehicle (192 wells) and positive control (1 OFIM Ebselen, 192 wells) from a single 384 well plate are shown. The Z' score for this assay was 0.51 with signal to noise (S/N) and signal to background (S/B) ratios of 12.8 and 2.9 respectively.
Figure 3 shows example spectrum library screening data for one 384 well plate. GFP fluorescence versus well number. Wells 1-24 and 360-384 contain alternate positive (1 Op.M Ebselen) and negative (vehicle) controls. Dotted line represents the average +3SD of the negative controls and all compounds above this line are counted as hits'.
Some wells show reduced fluorescence, most likely due to toxicity.
Figure 4a shows overlaid concentration response curves for all 46 hit compounds. Note that the profiles for many of the hit compounds follow a bell shaped dose response curve with reduced fluorescence due to toxicity at higher concentrations and have a narrow window of ARE induction. Figure 4b shows examples of compounds with minimal toxicity at high concentrations or a broad window of ARE induction, note most of these compounds showed some toxicity at I OOpM.
Figure 5 shows pharmacophore model and alignment of compounds from N Primary screen (compounds with ECSO <51sM) and B/ Astrocytic oxidative stress assay in 1321 NI cells (compounds with EC5O c31.xM). AromaticfHydrophobic feature in green, Hydrogen Bond Acceptor feature in blue. For (B) a basic pharmacophore emerges with two hydrophobic features associated with a hydrogen bond acceptor. This is consistent with known NRF2 activators which may act by nucleophilic attack of sulphydryl groups on KEAPI, the cytoplasmic NRF2 regulator.
Figure 6 shows the results of ARE reporter assay in C6 astrocyte cell line for 17 best hit compounds and S [+J apomorphine. Overall the response is similar to that seen in the CHO cell line and both R[-J and S[+J apomorphine induce the Nrf2-ARE pathway to a similar degree suggesting that this activity is unrelated to the dopamine agonist activity of R[-j apomorphine The response in the N5C34 cell line was substantially reduced or non-existent for the majority of compounds (not shown).
Figure 7 shows quantitative RT-PCR analysis for NRF2 regulated genes in the rat CS astrocyte cell line (C6 astrocytes, Figure 7a and Figure 7b) and primary mouse astrocytes (Figure 7c and Figure 7d) following 24h treatment with Andrographolide (Andro) and SN] Apomorphine (Apo 5) at EC and EC90 concentrations, determined in C6-4xARE-TK reporter cells. Mutliplex PCR was used to interrogate expression levels of 9 genes of interest including Nrf2 itself following drug treatment in CS cells. Only two genes, Haem oxygenase 1 and WOOl, showed statistically significant changes in gene expression, shown in (a) and (b) respectively. Standard quantitative RT-PCR of these two genes in primary mouse astrocytes under the same conditions were also performed (c) and (d). Asterisks indicate significant difference in DMSO control by one way ANOVA.
Figure 8 shows NRF2 inducers protect motor neurones (MN) in primary mouse astrocyte/MN co-cultures from menadione stress. Co-cultures were pre-treated for 24h with S[1 Apomorphine (Apo 5) and Andrographolide (Andro) at their EC50 and EC90 concentrations respectively, as determined in rat CS 4xARE-TK reporter cells. The co-cultures were then challenged for 6 hours with I OpM menadione to induce oxidative stress. In DM50 control cells an approximately 25% reduction in motor neurone number was observed which was not seen in wells treated with either drug Figure 9 shows immunoflourescence staining for Haem Oxygenase 1 in primary mouse astrocytes following treatment with Nrf2 Inducers at EC50 and EC90 concentrations. Area and staining intensity were quantified using Image J and used to calculate a staining index.
Figure 10 shows total Glutathione levels in primary mouse astrocytes (Figure 1 Oa) and in conditioned media collected from primary mouse astrocytes (Figure lOb) following treatment with Nrf2 lnducers at EC50 and EC90 concentrations. Total glutathione levels were measured using standard methods following 24h pre-treatment with Nrf2 inducers.
All treatments significantly increased glutathione levels in astrocytes relative to DMSO control and the EC90 concentrations of both drugs significantly increased extracellular glutathione levels (*p values <0.005). Data are average of three independent experiments Figure 11 shows quantitative RT-PCR analysis for the Nrf2 regulated genes NQOI (a) and Haem oxygenase I (HOXI) in primary mouse astrocytes overexpressing G93A mutant SODI following 24h treatment with Andrographolide (Andro) and S[i-] Apomorphine (Apo 5) at EC50 and EC93 concentrations. Asterisks indicate significant difference from DM80 control by one way AWOVA.
Figure 1 2A shows the in vivo pharmacokinetic time course for S[+] Apomorphine (Apo 5) and Figure 12B shows the corresponding PK parameters in mice (n=3 or 4 per time point). Figure 12C shows the QRT-PCR for HO-I and WOO-I in mice 6, 24 and 48 hours after a single subcutaneous injection of either 2.5 or 5.0 mg/kg S[+] Apomorphine (Apo 5).
DETAILED DESCRIPTION
Ce/I culture Chinese Hamster Ovary (CHO), NSC34 mouse motor neuronal cells, C6 (rat) and 1321 NI (human) astrocytic lines were routinely maintained in UMEM supplemented with 10% FBS and penicillin/streptomycin. The TK-EGFP reporter construct consists of a I 23bp thymidine kinase promoter inserted in the multiple cloning site of pEGFP (Clontech) and the ARE-TK-EGFP also contains four repeats of a 41 bp GST ARE motif (TAGCTTGGAAATGACATTGCTAATCGTGACAAAGCAAC1Tr) (SEQ ID NO:l) 3' to the TK promoter. These plasmids were transfected into CHO, C6 and 1321 NI cell lines using Lipofectamine 2000 (Invitrogen) and following 10-14 days of selection in 0.5mg/mi 0418 they were expanded and selected for basal eGFP expression using fluorescence activated cell sorting (BD, FACSAria) with two sequential cell sorts for each cell line.
These mixed populations of stable transfectants with basal eGFP expression were used in subsequent assays and designated 4xARE-TK-GFP for the ARE containing line and TK-GFP for the control cell line. The NSC34 cells lines were transfected with GG3A mutant SODI and stably transfected single cell clones were isolated by selection in 2SOpgIml G418 and cloning by limiting dilution.
ARE reporter assay -Spectrum llbrary screen validation In order to screen the spectrum library of 2000 small molecule drugs and natural products the TK-GFP CHO ARE reporter cell line was subjected to a I score assay in a 384 well plate (Greiner Bio-one, p.Clear, black) using a range of plating densities (5- 20x1 04/well plated 24 hours prior to assay) and different media. Alternate wells were incubated with 10gM of Ebselen and vehicle (0.1 % DM50) for 24 hours followed by replacement of media with PBS containing 0.3gM of ethidium homodimer-1 (EthDl).
This concentration of Ebselen represents an approximate EC90 for this drug. GFP fluorescence (ARE induction) was then measured at Ex4a5flfl/Emsaoflm using a Fusion universal plate reader (Packard Bioscience). The z score was calculated as follows 7= 1-(351Y+ 35D) (Avet -Aye) where SD4 standard deviation of positive control wells 3SD standard deviation of negative control wells Ave4 = Average fluorescence reading of positive control wells Ave = Average fluorescence reading of negative control wells Signal to noise (S/N Avet/SD4) and signal to background (S/B = Ave4/Ave) ratios were also determined for the different assay conditions. Acceptable Z' scores were >0.5.
For the library screen, cells were plated at a density of 20x104 in normal DMEM media containing 10% FBS on day -1 and on day 0, cells were incubated for 24 hours with drug in serum free media. Media was removed by hand and replaced (1 compound/well) with the Spectrum library diluted to 10gM in 0.1% DMSO using a 0-bot liquid handling system (Genetix, New Milton, UK). The media was removed after 24 hours and replaced with the same volume of PBS containing 0.3gM EthDl. GFP fluorescence (ARE induction, Ex4B5nm/Em53onm) and Eth Dl fluorescence (toxicity Ex530 nm/Em645 nm) were then measured using a Fusion universal plate reader (Packard Bioscience) The TK-.GFP CHO ARE cell line was screened twice in a single point assay and the control TK-GFP CHO cell line was screened once to eliminate false positives. Figure 1 shows a schematic representation of the NRF2-ARE assay.
ARE reporter assay -determination of EC50 Confluent cultures of 4xARE-TK-GFP CHO or TK-GFP CHO expressing cells in 96 or 384-well tissue-culture plates were treated with drug (0.01-1 00DM in triplicate) or vehicle (0.1-1% DM50) in FCS-free DMEM in triplicate for 24 hours. The media was removed and replaced with the same volume of PBS containing 0.3pM EthDl. GFP fluorescence (ARE induction, Ex4s5nmIEmssonm) and Eth Dl fluorescence (toxicity Ex5 nrn/Em645 urn) were then measured using a Fusion universal plate reader (Packard Bioscience). Non-linear regression was used to fit a sigmoidal dose-response curve on a semi-Log plot to calculate the EC50 using GraphPad Prism (GraphPad Software). The reporter assay was conducted in a similar fashion in C6 and 1321N1 astrocyte cell lines stably transfected with the 4xARE-TK-GFP and TK-GFP constructs except that Eth Dl was added directly in the media and read prior to washing the cells once and reading the DCF signal.
Oxidative stress assay A simple oxidative stress assay was used to determine whether preconditioning with NRF2-ARE inducing drugs could protect against a subsequent oxidative stress insult (serum withdrawal). The NSC34, C6 and 1321 Ni cells were plated in 96 well tissue culture plates to achieve 30% confluency. They were incubated with drug in triplicate wells as a 9 point titration (100DM to lOnM) for 24 hours. Cell density was observed to ensure no significant toxicity or growth inhibition occurred. Media was then replaced with serum free, phenol free media for 5 hrs. Dichlorofluorescein (DCF) and ethidium homodimer (EthDl) (Molecular Probes, Paisley, UK) was added to the cells to a final concentration of S 1iM and 0.3 RM, and DCF and EthDl fluorescence was read at Ex485 nrnIEms3o urn, Ex530 urn/Em645 urn respectively after 1 hour. Cell survival assay was then performed on the cells as protection is measured as % reduction in DCF signal, therefore data points were excluded where a decrease in cell number was measured.
Cell viability assay The method used was essentially as described previously in the art. Briefly methylthiazolyldiphenyl-tetrazolium bromide (MU) was added to the cells and a blank well to a final concentration 0.5mg/mI and incubated at 37 °C for 1-3 hrs depending on the cell line used. Cells and reaction product were solubilised in 20%SDS/50%DMF for 1 hr with shaking at room temperature before reading the absorbance at Ex5 nm-Basal oxidative stress assay in G93A SODI expressing NSC34 cells (ALS cellular S model) The G93A SODI expressing NSC34 cells were plated in 96-well tissue-culture plates in phenol red-free DMEM containing 10 % FBS until 30-40 % confluent They were then incubated with drug at 0.01, 0.1, 1, 1 OriM or vehicle (0.1% OMSO) in triplicate wells for 24hours. Cytosolic reactive oxygen species levels were measured using DCF and EthDl as in the oxidative stress assay.
Primary mouse motor neurone/astrocyte co-cultures Mouse glial cultures were established from C57B116 cortices from I -2 day old pups.
Cortices were dissected out and cells dissociated by trituration following incubation in DNasel, collagenase and trypsin. The incubation and trituration steps were repeated to ensure complete dissociation of cells. Cells were plated at 45,000 cellslcm2 in DMEM containing 10 % FBS, 100 U/mI penicillin, and 100.tglml streptomycin. After 24 hours1 cultures were washed in PBS and grown for 2 -3 weeks until confluent, changing the medium once a week. Confluent glial cultures were enriched for astrocytes by shaking and mild trypsinisation. Enriched astrocytes were grown to confluency and plated onto coverslips coated with poly-D-ornithine (1.5 mg/mI; Sigma, Poole, UK) at 40,000 cells/cm2, and grown for I week.
Primary spinal cord motor neurons (MN5) were cultured from El 3.5 wild-type C7B116 mouse embryos. Briefly, spinal chord preparations were dissociated by trituration following incubation in trypsin and DNasel. MNs were then isolated by density gradient centrifugation. Co-cultures were established by plating MNs at 8000/cm2, on astrocytes, in Neurobasal medium supplemented with 1% B27, 2% horse serum, 50 mg/mI streptomycin, 50 U/mI penicillin, 0.5 mM L-glutamine, 25 mM glutamic acid (all from lnvitrogen, Paisley, UK), Ing/ml BDNF, 10 ng/ml CNTF, and 100 pg/mI GDNF (all from R&D Systems, Abingdon, UK).
When co-cultures were established for 2 weeks, neuroprotection assays were performed by 24 hrs exposure to drug or vehicle followed by a 6 hour 10 micromolar menadione oxidative stress or glutamate. Following stress treatment, coverslips were washed 3 times, fixed and permeabilised to selectively stain motor neurons with 5M132 (Covance).
Total MNs were counted by fluorescent microscopy in a 1.5cm2 area per coverslip.
Minimum three repeats in triplicate were performed per condition. Both vehicle and drug treatments were counted before and after stress treatment, and results were statistically analysed by 2 way anova using Bonferroni post tests.
Total glutathione assay Primary astrocytes were grown to confluency in 24 well plates and then treated with drug (or 0.05% DM50 vehicle) in phenol red-free DMEM containing 10 % FBS and penicillin/streptomycin for 24 hours. Conditioned medium was collected and astrocytes were then washed in ice-cold PBS before addition of 250 p1fwell of sulphosalicylic acid (SSA, 5 % (wlv)). Plates were frozen at -80°C and thawed at 37°C, twice, and then incubated at 4°C for 15 minutes. The supernatant was removed and centrifuged at 13,000 x g for 5 minutes. Conditioned medium samples were incubated at 80°C for 15 minutes and then centrifuged at 13,000 x g for 5 minutes. Samples were either used immediately or stored at -80°C. Reaction mixture (150 pilwell; 100mM potassium phosphate buffer, pH 7.0, 1mM EDTA, 6 Units/mI glutathione reductase, 1.5 mg/ml 5,5'-dithiobis(2-nitrobenzoic acid)) was added to 10 p1 of each sample or glutathione standard (0 -50 p.M reduced glutathione) in a 96-well plate and incubated at room temperature for 5 minutes before addition of 50 j.tlIwell of NADPH solution (0.16 mg/mI).
A412nm was measured every minute for 15 minutes and the total glutathione concentration (GSH + GSSG) was calculated from initial rates. Samples were tested in triplicate.
In silico analysis In order to select drug-like molecules for further screening, Pipeline Pilot (SciTegic, London, UK) was used for in silico analysis. The molecular polar surface area (mPSA) was calculated for all 2000 molecules from the SPECTRUM collection as a crude measure of likely CNS penetrance {Ertl, 2000 #834} and a Lipinski Filter was also applied to determine which molecules were most drug-like. This filter applies the rule of five' which selects compounds with a cLogP c5, molecular mass <500, <5 hydrogen bond donors (OH+NH count) and <10 hydrogen bond acceptors (0 plus N atoms).
Quantitative RT-PCR Mutiplex PCi was used to detect changes in expression levels of target genes in the 1321 NI astrocyte cell line following treatment with Andrographolide and S(+) Apomorphine at EC50 and EC concentrations as determined in the NRF2-ARE reporter assay in C6 cells. The GenomeLabTM GeXP Genetic Analysis system (Beckman Coulter) was used to identity changes in gene expression in a multiplexed reaction for 9 genes of interest (Hmoxl, Fthl, Keapi, OcIc, Nfe2I2, Gsr, Nqol, Sqstml and EphXl) and three housekeeping genes (1 8s GAPDH and ACTB) using the following primers, outlined in
the Table 1 below:
Primer Produ Left Primer Sequence 5'-Left Right Primer Sequence -Right Tm Name ct Size 3' Tm AAGA000TMGACCGCC TGTGTGTGAGGGACTC Hmoxi 135 59.983 60.299 1TC (SEQ ID NO:2) TGGTC (SEQ ID I'40:3)
CCTGAATGAGCAGGTGA CAGGGIGTGCTTGTCA
Fthl 152 59.685 59.873 AATC (SEQ ID NO:4) AAGA (SEQ ID NO:5)
ACGACGTGGAOACAGAG AAAAGTGTGGCCATCG
Keapl 159 60.315 60.14 ACC (SEQ ID NO:6) TAGC (SEQ ID NO:7)
AAAGGCCTCTAAGCCAG GCAGATAGTGGCCAAC
GcIc 173 59.848 60.142 ACC (SEQ ID NO:8) TGGT (SEQ ID NO:9)
AGACAGCCGCATCITCT TTGATGGCMCAATGTC
Gapdh 180 60,02 60.376 TGT (SEQ ID NO:10) GAG (SEQ ID NO:11)
AMGGGCTACCACATCC CCTCCMTGGATCCTC
lBS 192 59.993 59.887 MG (SEQ ID NO:12) GTTA (SEQ ID NO:13) CACCATAGAGGAGGACA CAGTGAG000ATCGAT Nfe2I2 212 60.287 60.072 TOG (SEQ ID NO:14) GAGT (SEQ ID NO:15) CGCCCATAGACT1'TTTG CAmcGTcTrCCTCGT Oar 241 59.702 60.111 AGG (SEQ ID NO:16) GOT (SEQ ID NO:17) ACCTTGCmCCATCAC TCTCCAGACGCTTCTIC Nqoi 251 59.973 60.533 CAC (SEQ ID NO:18) CAC (SEQ ID NO:19) 1TGCTGACAGGATGCAG M000TGTAAMCGCA Actb 273 60.136 59.383 MG (SEQ ID NO:20) GCTC (SEQ ID NO:21) Sqstml
TGGCCATGTCCTATGTG MGGGGTTGGGAMGA
(vi and 296 59.924 60.298 AAA (SEQ ID NO:22) TOAG (SEQ ID NO:23) v2) Ephxl (vi 000CTTGACATCCACYF CCCGCCTTGMTGTAG 349 59.943 60.318 and v2) CAT (SEQ ID NO:24) MITT (SEQ ID NO:25) Those shown in bold flank intronic sequences, preventing genomic background. The RT reaction mixture was assembled in a 96 well microplate as follows 3gl DNase/RNase Free H20, 4jsl RT Buffer 5X, 2.d RT Rev Primer Plex Reverse Transcriptase, 5g1 KANr RNA, 5pi Sample RNA (20 ng/pL) to gve a total reaction volume of 2OjiI. The samples were then incubated as follows, 48°G 1 minute, 37°C 5 minutes, 42°C 60 minutes, 95°C 5 minutes, 4°C Hold. The PCR reaction mixture was assembled as follows in a 96-well sample microplate; 4.Ogl PCR Buffer 5X, 4.Ogl 25 mM MgCl2 (Abgene), 2.Ogl PCR Fwd Primer Pie; 0.7tl Thermo-Start DNA Polymerase, (ABgene AB-09081A), 9.31.Li cDNA Samples from RI reaction. The PCR was run as follows, (1) 95°C 10 minutes, (2) 94°C 30 seconds1 (3) 55°C 30 seconds, (4) 70°C 1 minute, (5) Repeat steps 2-4 for an additional 34 cycles, (6) 4°C Hold. On the basis of preliminary experiments reverse RI primer concentrations were optimised to allow detection of all products in a single reaction. PCR reaction products were diluted in SLS (sample loading solution) and separated on the GeXP capillary electrophoresis unit and the data analysed using the GeXP software and Microsoft Excel data package to give a fold change in expression relative to GAPDH and ACTB.
Standard QPCR QPCR was carried out as previously described in the publication Wood-Allum et al 2005 with the essential difference that RNA was isolated from primary astrocytes or the astrocyte cell line using the RNeasy kit (Qiagen) following manufacturers' instructions.
cDNA was prepared using Superscript II (Invitrogen) driven by oligo dl following manufacturers' guidelines. Quantitative RT-PCR. (Q-PCR) was performed using 25 ng of cDNA, lx SY13R Green PCR Master Mix (Stratagene) and optimised concentrations of forward and reverse primers (Table 1), in a total volume of 20 p1 and in triplicate for each sample. Following denaturation at 95 °C for 10 mm, products were amplified by 40 cycles of 95°C for 15 s and 60 °C for 1mm, on an MX3000P Real Time PCR System (Stratagene). To ensure amplification of a single product the dissociation curve was produced for each amplification. The relative levels of the genes of interest in the samples was determined by normalizing the level of expression to that of gapdh in each of the samples and by using the ddCt calculation (SYBR Green PCR mix and RT-PCR protocol, Applied Biosystems). Overall relative concentrations of the genes of interest were calculated following drug treatment by normalising the data to the cells grown in the vehicle (DMSO).
Statistical analysis For the Spectrum library screen, each 384 well plate contained 32 wells incubated with vehicle only (0.1% OMSO in media). The average fluorescence reading and standard deviation of these wells was calculated on a plate by plate basis. Hits were classified as having a GFP fluorescence value greater than the vehicle average plus three standard deviations. Toxicity was defined in the same way (i.e. EthDl fluorescence value greater than the vehicle average plus three standard deviations).
EXAMPLE I
The 4xARE-TK-GFP and TK-GFP reporter cell lines were tested for their response to the known ARE inducers tert-Butyl Hydroquinone (tBHQ) and the flavonoid EGCG at a range of concentrations. The compounds were applied in triplicate to confluent cells in 96 well plates in serum free medium for 24 hours and the induction of GFP measured in a fluorescence plate reader. Both compounds induced GFP expression in a narrow window, with EGCG peaking at 1 001.xM and tBHQ peaking at 1 0j.zM (Fig 2a). At concentrations higher than this peak expression, both compounds showed signs of toxicity by direct observation (cell loss) or increased Ethidium Homodimer fluorescence.
No increase in fluorescence was seen in the control TK-GFP cell line (not shown).
EXAMPLE 2
In order to screen the SPECTRUM collection of 2000 molecules the reporter assay was scaled down to a 384 well-plate format. To assess the suitability of the assay for library screening, a Z' score calculation was performed by treating alternate wells with vehicle (0.1% DM50) and 10 pM Ebselen as a positive control (see calculation in Methods).
We have shown Ebselen gives a robust concentration response curve in this assay. The calculated Z' score was 0.51 (Fig 2b) which is acceptable for library screening. In addition, signal to noise (SIN) and signal to background (S/B) ratios were acceptable at 12.8 and 2.9 respectively. The library was subsequently screened at a single concentration per compound of 10gM. Drug library dilutions and plating were carried out by a Q-BOT liquid handling system and both the 4xARE-TK-GFP reporter cell line and TK-GFP control cell line were tested for their response to the compounds. An example set of data for the ARE-TK-GFP cell line from a single 384 well plate is shown in Figure 3. Hits were identified as having data points more than three standard deviations above the background level, which was the average value of 24 wells treated with vehicle (0.1% DM50) only. Hit compounds were checked to see if they generated a response in the control cell line due to either non-specific activation of transcription or autofluorescence of the compounds. In addition, any compounds showing signs of toxicity by enhancing ethidium homodimer fluorescence were excluded. The library screen was repeated with the 4xARE.-.TK-CHO cell line only and compounds which emerged as hits from both screens were taken forward for further assessment. A total of 46 compounds were identified on this basis. The next step was to determine the compound concentration required to give a 50% response (EC50) for these 46 hit compounds. Each compound was subjected to a 7 point concentration response curve in duplicate wells. Many compounds showed a bell shaped dose response curve similar to that seen for standard ARE inducers such as tBHQ and EGCG, due to toxicity at higher concentrations. Figure 4a shows all 46 dose response curves in the first assay. The majority of compounds exhibit a bell-shaped dose response curve with toxicity at higher concentrations and many also have a very narrow window of ARE induction. Figure 4b shows a set of compounds with enhancement of reporter expression over a broader concentration range (>1 log unit) or with minimal toxicity at higher concentrations. The concentration response curves for all 46 hits were repeated and the average EC50 and average maximum fold induction of GFP fluorescence measured. The lowest concentration which caused a toxic response was also noted and the data are summarised in Table 2 (presented herein after), together with a brief description of the known bioactivity of these compounds. The lowest dose at which toxicity was observed is also included in the table. Compounds are ranked according to activity in the reporter assay. The most potent ARE inducer was the natural product andrographolide, the only compound with a sub-micromolar EC50 (740 nM), this compound comes from the natural product Andrographis paniculata, and is used widely in Chinese and Indian herbal medicine. Of the 26 other natural products, a further two have been used in man; securinine, a GABM receptor antagonist and CNS stimulant and isoliquiritigenin, a component of liquorice root which is an aldose reductase inhibitor. The remaining 19 products were synthetic small molecules or derivatives and of these a total of six molecules were approved drugs. Two alkylating anti-neoplastic drugs (pipobroman and mechiorethamine) a dopamine agonist (apomorphine hydrochloride), a topical skin whitener (hydroquinone), a loop diuretic (ethacrynic acid) and a vasodilator (isoxsuprine hydrochloride). One of the synthetic small molecules had reached phase three clinical trials in stroke (Ebselen).
EXAMPLE 3
The effects of Nrf2-ARE inducing hit compounds on oxidative stress induced by serum withdrawal in motor neuronal and astrocytic cells was investigated. Since the activation of this pathway may vary depending on the cell type, we then went on to screen how well these hit compounds could protect a motor neuronal cell line (N5C34 cells) and rat (C6) and human (132911) astrocyte cell lines from oxidative stress induced by serum withdrawal. The cell lines were pre-treated with hit compound at a range of concentrations for 24 hours to activate the NRF2-ARE pathway. The compound was then removed and the cells subjected to a six hour serum withdrawal to induce oxidative stress. The degree of oxidative stress was measured using dichlorofluorescein (DCF) fluorescence and the degree of protection is shown in Table 3 as percentage reduction in DCF fluorescence for each of the three cell lines. Where it was possible to fit a curve, the concentration required to give a half maximal effect (IC50) is also quoted. In general, hit compounds were more likely to show protective effects in the astrocyte cell lines than in the motor neuronal cell line. Table 3 (presented herein after) gives the assay results for all compounds, ranked by activity in the motor neuronal cell line. Only 9/46 compounds redubed the oxidative stress DCF signal induced by serum withdrawal in NSC34 cells, 18/46 compounds had no effect in this assay and the remaining 17 compounds were pro-oxidant in this assay. ln other words as opposed to decreasing the oxidative stress caused by serum withdrawal they contributed to it and increased DCF fluorescence. In contrast only one compound was pro-oxidant in the 1321 NI astrocyte cell line and 29/46 reduced the DCF signal by 30% or more. For the C6 cell line no compounds were pro-oxidant and 32/46 compounds reduced the DCF signal by 30% or more.
EXAMPLE 4
In order to rationalize the biological results obtained, a general pharmacophore for the compounds reported in Table I was investigated, using the Pharmacophore Elucidator implemented in MOE (Molecular Operating Environment) [Molecular Operating Environment (MOE 2007.09). Chemical Computing Group, Inc. Montreal, Quebec, Canada http://www.chemcomp.coml. Considering the 24 molecules with an EC50 of less than 10pM for induction of the NRF2-ARE pathway in the CHO 4x ARE-TK cell line (Table 2), upon alignment, 22 of them presented two common features: an aromatic/hydrophobic moiety and a hydrogen bond acceptor moiety (Figure 5a). It should be noted that EGCG also possesses these structural features and positively matches the calculated pharmacophore. Furthermore, when the 1321 Ni astrocyte oxidative stress assay results (Table 3) were used for constructing the pharmacophore with a 3 pM activity threshold, one additional common aromatic/hydrophobic feature was identified (Figure 5b). This is consistent with known Nrf2 activators which may act by electrophilic attack of suiphydryl groups on KEAPI, the cytoplasmic Nr12 regulator.
These preliminary modelling data can be used to understand the structural requirements of potential activators of the Nrf2-ARE pathway and could be taken into consideration in the design of novel structures for this class of compounds.
EXAMPLE 5
The physical/chemical properties of the compounds were assessed. In addition to screening the compounds for functional effects in relevant cell types we also used Pipeline Pilot, a chemi-inforrnatics programme to calculate chemical/physical properties, also shown in Table 3. ALogP (log of the partition coefficient in octanol/water) and Molecular polar surface area (mPSA) are different measures of the lipophilicity of the compounds and allow crude prediction of likely CNS penetrance. For CNS penetrance an AlogP less than 4 but greater than 1, and a mPSA below 100 (ideally 80) are optimal for passive diffusion across the BBB.
In addition, the Lipinski filter was used to identify the non-drug like molecules and four were excluded-(swietenolide-3-acetate, endecaphylin X, lobaric acid and euphol acetate).
To filter out unwanted molecules only those with a protective or neutral effect in the NSC34 oxidative stress assay (Table 3) were selected and known cytotoxic molecules excluded (pipobroman, chiordane, alachlor, propachlor). Applying the AlogP and mPSA criteria to the remaining 22 molecules left 17 molecules for further investigation. These molecules are highlighted in bold in Table 3 and are designated the best hit' molecules.
In addition the minimum dose at which toxicity was observed is shown. NA, not applicable (insufficient data, no concentration response or no inhibition). For Table 3 compounds are sorted by protective capacity in the NSC34 cell line. ARE inducers were far more effective at protecting the astrocyte cell lines (1321 Ni and 06) from oxidative stress compared to N5C34 cells.
EXAMPLE 6
NRF2-ARE Inducing activity of the best hit compounds in neuronal and astrocytic cell lines was investigated. In order to determine whether the differences in protection in astrocytic and motor neuronal cell lines was due to differences in the degree of activation of the NRF2-ARE pathway in these cell types, the NRF2-ARE reporter construct was stably expressed in both the astrocytic (06) and motor neuronal (NSC34) cell lines. The 17 best hit molecules were then screened in each cell line. We also screened the S [+1 enantiomer of apomorphine as it can exist as either an R[] or S[+} enantiomer. The R[�] enantiomer has dopamine agonist activity whereas the S[+] enantiomer has lost this activity and so we wanted to determine whether it retains NRF2-ARE inducing activity. The results for the C6 reporter cell line are shown in Figure 6. In general activation of the NRF2-ARE pathway in the C6 cells was similar to that seen in the CHO cell line. The NSC34 reporter cell line showed minimal if any activation with the same set of concentration response curves suggesting the underlying cause for greater protection against oxidative stress in astrocytic cell lines versus the NSC34 cells seen earlier was due to a much more robust activation of the NRF2-ARE pathway in astrocyte cell lines, in addition, the S[+1 enantiomer of apomorphine was equally potent in terms of NRF2-ARE activation compared with the R[+] enantiomer indicating that this activity is unrelated to agonism of dopamine receptors as the S[+} enantiomer is not a dopamine agonist.
It was an objective to identify molecules with potential to be fast-tracked for clinical testing in Motor Neurone Disease. One of the key criteria in this regard was that the molecules have a history of use in man albeit not for MND. Of the 17 best hit molecules, two had a history of use in man as natural products (securinine, andrographolide) and one was a currently approved drug used for the acute rescue of off' episodes in Parkinsons Disease (apomorphine hydrochloride as the R enantiomer). In addition, hydroquinoine has a history of use, although as a topically applied skin whitener. Of these drugs andrographolide and the S[+1 enantiomer of apomorphine were selected for further assessment as securinine has well described convulsant properties in mice and man and the lack of dopamine agonism in the S[+J enantiomer of apomorphine was considered a distinct advantage.
EXAMPLE 7
Induction of ARE target gene expression in C6 cells and primary mouse astrocytes by andrographolide and S[+1 apomorphine was investigated. To confirm that these preferred or lead' molecules were able to activate the NRF2-ARE pathway leading to target gene expression in astrocytes, a multiplex RT-PCR assay was developed on the GenomeLabTM GeXP genetic analysis system for 9 genes of interest, C6 cells were treated for 24h with Andrographolide and S[+] Apomorphine at EC50 and EC90 concentrations as determined in C6-4xARE-TK reporter cells. Only two genes, Haem oxygenase 1 and NQOI, showed statistically significant changes in gene expression, which was confirmed by standard quantitative RT-PCR (Figures 7A and 7B).
EXAMPLE 8
Since the lead NRF2 inducing molecules were able to increase expression of target genes in primary mouse motor neurones, co-cultures consisting of primary mouse motor neurones (MN) on an astrocyte feeder layer were exposed to an oxidative insult following pre-treatment with either Andrographolide of [S+J apomorphine (Figure 8). The co-cultures were then challenged for 6 hours with 10pM menadione to induce oxidative stress and motor neurones stained and counted. In DMSO control cells an approximately 25% reduction in motor neurone number was observed which was not seen in wells treated with either drug. These results indicate that NRF2 inducers protect motor neurones (MN) from oxidative stress in primary mouse astrocyte/MN co-cultures.
Standard quantitative RT-PCR of the NOOl and HO-i genes in primary mouse astrocytes under the same conditions also evinced significant increases in gene expression (Figures 7C and 7D). In order to confirm that changes in gene expression lead to concomitant increases in protein expression, primary mouse astrocytes were assessed for elevated levels of HO-i by immunofluoresence staining (Fig 9) which confirmed a dose-dependent enhancement of expression over that seen in DMSO treated cells. We also examined a functional effect of this enhanced anti-oxidant capacity by examining total glutathione levels under the same conditions in both the primary astrocytes themselves and in the media collected from treated astrocytes (Fig 10). This showed that glutathione levels were elevated in both the cells themselves and in media -providing one mechanism by which Nrf2 responsive astrocytes may protect neighbouring nonARE responsive MNs from oxidative challenge.
EXAMPLE 9
In an attempt to rationalize the biological results obtained, we have tried do identify a general pharmacophore for the compounds reported in Table 2, using the Pharmacophore Elucidator implemented in MOE (Molecular Operating Environment) Molecular Operating Environment (MOE 2007.09). Chemical Computing Group, Inc. Montreal, Quebec, Canada http://www.chemcomp.com]. Considering the 24 molecules with an ECSO of less than 10pM for induction of the NRF2-ARE pathway in the CHO ARE-TK cell line (Table 2), upon alignment, 22 of them presented two common features: an Aromatic/Hydrophobic moiety and a Hydrogen Bond Acceptor moiety (Figure 4a). It should be noted that EGCG also possesses these structural features and positively matches the calculated pharmacophore. Furthermore, when the 1321 NI astrocyte oxidative stress assay results (Table 3) were used for constructing the pharmacophore, using a 3 pM activity threshold, one extra common Aromatic/Hydrophobic feature was identified (Figure 5b). This is consistent with known NRF2 activators which may act by nucleophilic attack of sulphydryl groups on KEAPI, the cytoplasmic NRF2 regulator.
These preliminary modelling data can be used to understand the structural requirements of potential activators of the NRF2-ARE pathway and could be taken in consideration in the design of novel structures for this class of compounds.
EXAMPLE 10
Since previous work had demonstrated an attenuated Nrf2 response in motor neuronal cell lines expressing mutant SOD and in post mortem material in astrocytes from familial human SODI cases we investigated whether our lead inducers could still activate Nrf2 in astrocytes expressing G93A mutant SODI. It was first determined whether the Nrf2 regulated genes NAD(P)F-tquinine oxidoreductase (NQOI) and heme oxygenase 1 (HOXI) could be induced in primary mouse astrocytes from G93A mutant SODI transgenic mice (Figure 11). Quantitative RT-PCR demonstrated a significant increase in transcripts for NQOI and HO1 following a 24 hour pre-treatment with Andrographolide and S[+] Apomorphine at their EC90 concentrations (Figure II).
EXAMPLE II
Figure 12 shows the in vivo pharmacokinetics and pharmacodynamics of [5+] apomorphine in male C57B1/6 mice. Following a single intravenous dose of [5+] apomorphine levels of the compound were detected in plasma, brain and cerebral spinal fluid (Figures 1 2A and I 2B) and following subcutaneous doses there was significant induction at 24 hours post dose of HO-I and NQO-1 transcriptsas determined by QRT-PCR (Figure 1 2C). Accordingly, [8+1 apomorphine can lead to a prolonged increase in expression of anti-oxidant enzymes through transcriptional activation.
These data demonstrate that several compounds have been identified that are NF2-ARE pathway activators in vitro and that they may also activate this pathway in vivo.
TABLE 2
ARE reporter assay Average Lowest fold GFP Average dose Name Bloactivity and/or source induction at EC5O with maximal (pM) toxicity response -(pM) Andrographolide (diterpenoid) Natural product used in Indian and Chinese Medicine 3.61 0.74 100 deoxygedunin (triterpenoid) Similar to known anti-malarial 1.82 1.15 30 Propachlor pesticide, high dose neurotoxic 3.25 1.49 100 Securinine GABAA receptor antagonist, alkaloid, CNS stimulant 3.00 2.12 30 Dalbergione Natural product 2.09 2.47 30 Isoliquiritigenin Licorice flavanoid, aldose reductase inhibitor 2.58 2,52 30 4 acetoxyphenol Natural product, antioxidant 2.78 2.91 100 3 acetoxypregn-16-en-12,20-dione Natural product derivative 3.14 2.98 30 2 methyl gramine Natural product derivative 2.27 3.00 100 hydroquinone (1,4 dihydroxybenzene) skin whitener, antioxidant 2.95 3.07 100 Alachior herbicide, high dose neurotoxic 3.50 3.29 100 kinetin riboside Purine derivative 1.97 3.39 30 Deacetylgedunin Natural Product 3.05 3.50 30 sappanone A trimethyl ether Natural product derivative 1.99 3.50 100 swietenolide 3 acetate possibly natural? Liminoids are antifungal 2.82 3.50 100 4 hydroxychalcone flavonol, glutathione reductase inhibitor, plant polyphenol 1.88 3.53 None Ebseten Antioxidant 2.81 4.00 100 Citrinin Natural product, antibacterial 3.26 4.10 30 Mechioroethamine antineoplastic, alkylating agent 2.25 5.50 30 Isoxsupdne hydrochloride Vasodilator 1.52 8.70 100 313 hydroxydeoxydesacetoxy-7-oxogedunin Natural Product 2.55 8.71 30 Chlordane pesticide, high dose neurotoxic 1.56 9.18 100 3 methylorsellinic acid Benzoic derivative, Phospholipase A2 inhibitors 2.87 9.59 100 Pipobroman antineoplastic, alkylating agent 3.21 10.0 100 Deoxyandirobin lactone (similar structure) Semlsynthetic 3.03 11.1 100 levulinc acid Natural product derivative 2.94 12.0 None Ethacrynic acid Diuretic 2.72 14.0 100 penicillic acid fungal toxin, antibacterial 2.64 14.9 100 4 methoxychalcone Natural product 2.13 15.0 100 3 methoxycatechol (similar to hydroquinone) Oncogenic in rats 3.28 15.4 100 Lobaric acid Natural product 2.53 17.0 100 epoxy(4,Scz)-4,5-dihydrosantonin anti parasitic, toxicity affects vision 2.91 18.2 100 nitrogenous glucoside miserotoxin (neu rotoxic and endecaphyllin X respiratory toxin) 2.15 18.8 None Benzyl isothiocyanate Natural product, antineoplastic, antibacterial, antifungal 1.81 24.0 None impotence drug and PD dopamine agonist, known CNS Apomorphine hydrochloride delivery 1.90 25.9 100 2,&dihydroxy-4-methoxytoluene 3.05 45.0 100 Parthenolide NSAID,HCV inhibitors 2.77 136 100 Isogedunin Natural Product 3.11 550 None methyl-7-deshydroxypyrogallin-4-carboxylate synthetic analog antioxidant 2.63 2054 100 4-Methoxydalbergione Natural product 2.11 7/11 100 Antiarol Natural product 1.64 1.5/23 100 Thymoquinone Natural product 2.58 1211.26 30 Euphol acetate Natural product 2.20 99? 100 Retusoquinone Natural product 2.04 NC/35? 100 one dose 3-alpha-hydroxygedinin Natural product 3.01 -effect 100 P123736W0.as filed
TABLE 3
N8C34 oxidative stress 1321N1 oxidative stress C6 oxidative stress assay Calculated assay assay properties Maximal % Maximal % Lowest Maximal Lowest dose reduction Lowest Name iC5O reduction in dose with lCSO reduction in with toxicity (pF) oxic7tive t(pM) ALogP mPSA stress (pM) stress stress Pipobroman NA 30 none 3.16 19 10 11 71 none 0.647 40.62 2,6-Oihydroxy-4-methoxytoluene -3.26 28 none NA 40 none 5.29 16 10 1.815 49.68 Apomorphine hydrochloride 0.174 25 10 1.7 79 none 1.69 62 none 3.498 43.7 4-Methoxychalcone NA 24 none 3.6 30 none 3.55 82 none 3.685 26.3 Securinine NA 20 none 18.3 55 none 0.645 67 none 1.448 29.53 Levulinic acid NA 15 none 3.31 58 none 7.69 62 none 1.652 54.37 3-Aipha-hydroxygedinin NA 14 10 NA 25 none 3.22 42 none 3.83 85.97 Penicillic acid NA 9 none NA 45 none 4.62 66 none 0.778 55.76 Deoxygedunin NA 7 10 1.41 25 10 2.19 61 none 4.217 82.81 Hydroquinone NA 0 none 3.38 80 none 7.07 67 none 1.346 40.46 Andrographolide NA 0 10 0.501 35 10 0.192 69 10 2.056 86.99 Deacetylgedunin NA 0 none 2.37 28 10 5.36 70 none 2.97 89.26 313 Hydroxydeoxydesacetoxy-7-oxogedunin NA 0 3 3.96 62 none 3.07 67 none 2.683 89.26 isogedunin NA 0 1 NA 10 none NA 3.349 95.34 Deoxyandirobin lactone NA 0 none 1.64 55 none 2.87 38 none 3.399 85.97 Endecaphyllin X NA 0 10 NA 56 none 3.15 40 none -9.6E-02 317.93 Parthenolide NA 0 1 28.4 47 none 3.11 81 none 2.923 38.82 Alachior NA 0 10 1.97 83 none 6.22 62 none 3.425 29.54 Propachior NA 0 10 0.829 50 10 0.645 74 10 2.415 20.31 Chiordane NA 0 3 NA 68 none 3.57 20 none 4.97 0 Epoxy(4,5a)4,5-dihydrosantonin NA 0 3 0.662 53 none 2.13 62 none 1.598 55.89 Kinetin riboside NA 0 3 0.246 25 3 3.84 40 10 -1.033 138.68 3 Acetoxypregn-1 6-en-I 2,20-dione NA 0 none 12.4? 75 none 5.34 50 none 3.213 60.44 Thymoquinone NA 0 none 11 25 none 0.014 32 none 2.288 34.14 Citrinin NA 0 none 0.338 28 none 20 23 none 0.928 83.83 Dalbergione NA 0 none 54 38 none 46 40 none 2.94 34.14 Swietenolide 3 acetate NA 0 0.3 NA 60 none 4.22 50 none 2.633 129.34 Isoliquiritigenin NA -6 1 1.78 50 none 28? 46 none 2.976 77.76 Mectiloroethamine NA -10 10 0.302 37 10 3.42 67 none 1.65 3.24 3-Methylorsellinic acid NA -12 0.3 3.26 53 none 3.1 56 none 1.948 77.76 3-Methoxycatechol NA -13 0.3 NA 39 none 1.2 46 10 1.329 49.68
Table 3 continued
N5C34 oxidative stress 1321N1 oxidative stress Cal culated C6 oxidative stress assay assay assay properties Maximal % Maximal % Lowest Maximal % Lowest dose reduction Lowest 1C50 1050 reduction in dose with 1050 reduction in with toxicity n dose with ALogP mPSA Name (pM) Oxidative toxicity (pM) Oxidativè (pM) (pM) Oxidative toxicity (pM) stress (pM) stress stress Isoxsuprine hydrochloride NA -16 none NA ? none 23 40 none 3.384 61.71 Methyl-7-deshydroxypyrogallin-4-carboxylate NA -23 0.3 NA 45 none 2.95 70 none 1.133 104.06 Retusoquinone NA -25 none 0 none 2.06 15 none 2.684 17.07 Lobaric acid NA -26 none 0 none 3.14 9 none 5.993 119.36 Benzyl isothiocyanate NA -28 none NA 8 none 3.47 27 none 2.596 44.45 Ethacrynic acid NA -30 10 3.01 25 none 3.01 31 none 3.964 63.6 2-Methyl gramine NA -30 3 11 36 none 32 36 none 2.342 19.03 Euphol acetate NA -43 none 18 38 none NA 0 none 8.498 26.3 4-Methoxydalbergione NA -44 none NA 43 none 2.5 14 none 2.627 43.37 Antiarol NA -50 10 NA 35 none 3.05 23 10 1.538 47.92 Sappa none A trimethyl ether NA -60 none 0.279 40 none NA 27 none 3.35 53.99 4-Acetoxyphenol NA -70 none 0.298 42 none NA 0 none 1.355 46.53 Ebselen NA -80 none 0.193 20 none NA 30 none 3.227 20.31 4-Hydroxyclialcone 3.459 37.29
GB0918626A 2009-10-23 2009-10-23 Treatments for neurodegenerative disorders Withdrawn GB2464813A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
GB0918626A GB2464813A (en) 2009-10-23 2009-10-23 Treatments for neurodegenerative disorders

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB0918626A GB2464813A (en) 2009-10-23 2009-10-23 Treatments for neurodegenerative disorders

Publications (2)

Publication Number Publication Date
GB0918626D0 GB0918626D0 (en) 2009-12-09
GB2464813A true GB2464813A (en) 2010-05-05

Family

ID=41426917

Family Applications (1)

Application Number Title Priority Date Filing Date
GB0918626A Withdrawn GB2464813A (en) 2009-10-23 2009-10-23 Treatments for neurodegenerative disorders

Country Status (1)

Country Link
GB (1) GB2464813A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9200046B2 (en) 2011-06-29 2015-12-01 Cornell University Reporter system for high throughput screening of compounds and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996017605A1 (en) * 1994-12-06 1996-06-13 Paracelsian, Inc. Use of andrographolide compounds to treat or prevent pathogenicity of diseases
WO2002009698A1 (en) * 2000-08-01 2002-02-07 Ashni Naturaceuticals, Inc. Compositions exhibiting synergistic inhibition of the expression and/or activity of cyclooxygenase-2
WO2005074953A1 (en) * 2004-02-03 2005-08-18 Universidad Austral De Chile Composition of labdane diterpenes extracted from andrographis paniculata, useful for the treatment of autoimmune diseases, and alzheimer disease by activation of ppr-gamma receptors
JP2007176934A (en) * 2005-11-30 2007-07-12 Kose Corp Platelet-activating factor acetylhydrolase function regulator
WO2008108825A2 (en) * 2006-10-10 2008-09-12 Burnham Institute For Medical Research Neuroprotective compositions and methods

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996017605A1 (en) * 1994-12-06 1996-06-13 Paracelsian, Inc. Use of andrographolide compounds to treat or prevent pathogenicity of diseases
WO2002009698A1 (en) * 2000-08-01 2002-02-07 Ashni Naturaceuticals, Inc. Compositions exhibiting synergistic inhibition of the expression and/or activity of cyclooxygenase-2
WO2005074953A1 (en) * 2004-02-03 2005-08-18 Universidad Austral De Chile Composition of labdane diterpenes extracted from andrographis paniculata, useful for the treatment of autoimmune diseases, and alzheimer disease by activation of ppr-gamma receptors
JP2007176934A (en) * 2005-11-30 2007-07-12 Kose Corp Platelet-activating factor acetylhydrolase function regulator
WO2008108825A2 (en) * 2006-10-10 2008-09-12 Burnham Institute For Medical Research Neuroprotective compositions and methods

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Environmental Toxicology & Pharmacology (2008); Vol 25, pp 321-328, "Amelioratory effect of Andrographis...", Neogy et al *
Journal of Pharmacology & Experimental Therapeutics (2004); Vol 308, pp 975-983, "Andrographolide reduces inflammation-related...", Wang et al *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9200046B2 (en) 2011-06-29 2015-12-01 Cornell University Reporter system for high throughput screening of compounds and uses thereof

Also Published As

Publication number Publication date
GB0918626D0 (en) 2009-12-09

Similar Documents

Publication Publication Date Title
US20110251230A1 (en) Therapeutics for neurological disorders
Melli et al. Alpha-lipoic acid prevents mitochondrial damage and neurotoxicity in experimental chemotherapy neuropathy
Zhang et al. p53 mediates mitochondria dysfunction-triggered autophagy activation and cell death in rat striatum
US10301630B2 (en) Methods and compositions for unsilencing imprinted genes
Huang et al. L-Ascorbate protects against methamphetamine-induced neurotoxicity of cortical cells via inhibiting oxidative stress, autophagy, and apoptosis
Wang et al. Resveratrol modulates angiogenesis through the GSK3β/β-catenin/TCF-dependent pathway in human endothelial cells
US11221329B2 (en) Treatment of neurological and neurodevelopmental diseases and disorders associated with aberrant ion channel expression and activity
Zhong et al. Inhibition of PDE4 by FCPR16 induces AMPK-dependent autophagy and confers neuroprotection in SH-SY5Y cells and neurons exposed to MPP+-induced oxidative insult
Liu et al. A novel inhibitor of homeodomain interacting protein kinase 2 mitigates kidney fibrosis through inhibition of the TGF-β1/Smad3 pathway
EP3262021B1 (en) Treatment method, compounds, and method of increasing trpv2 activity
Zhang et al. Isoliquiritigenin protects against blood‑brain barrier damage and inhibits the secretion of pro-inflammatory cytokines in mice after traumatic brain injury
Zheng et al. Metformin prevents peritendinous fibrosis by inhibiting transforming growth factor-β signaling
Zhang et al. Novel role for TRPC4 in regulation of macroautophagy by a small molecule in vascular endothelial cells
WO2013134546A1 (en) Methods and materials for treating cancer
SG189519A1 (en) TREATMENT OF MeCP2-ASSOCIATED DISORDERS
US11938135B2 (en) Compositions and methods for treating vascular Ehlers Danlos syndrome and associated disorders
WO2015120257A2 (en) Uses of gli1 in detecting tissue fibrosis
Yoshioka et al. Cyclic AMP‐elevating agents prevent oligodendroglial excitotoxicity
Zeng et al. EGCG protects against myocardial I/RI by regulating lncRNA Gm4419-mediated epigenetic silencing of the DUSP5/ERK1/2 axis
GB2464813A (en) Treatments for neurodegenerative disorders
Zhu et al. Bafilomycin A1 attenuates osteoclast acidification and formation, accompanied by increased levels of SQSTM1/p62 protein
Kang et al. KBH-A42, a histone deacetylase inhibitor, inhibits the growth of doxorubicin-resistant leukemia cells expressing P-glycoprotein
US8975293B2 (en) Epigenetic co-repressors of the gamma-globin gene and methods of using same
US9545396B2 (en) Method and pharmaceutical composition for inhibiting PI3K/AKT/mTOR signaling pathway
AU2021200051A1 (en) Method of treatment and compositions comprising a dual PI3K delta-gamma kinase inhibitor and corticosteroid

Legal Events

Date Code Title Description
WAP Application withdrawn, taken to be withdrawn or refused ** after publication under section 16(1)