Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 28(11): 115455, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32307259

ABSTRACT

Gamma-secretase modulators (GSMs) selectively lower amyloid-ß42 (Aß42) and are therefore potential disease-modifying drugs for Alzheimer's disease (AD). Here, we report the discovery of imidazopyridine derivatives as GSMs with oral activity on not only Aß42 levels but also cognitive function. Structural optimization of the biphenyl group and pyridine-2-amide moiety of compound 1a greatly improved GSM activity and rat microsomal stability, respectively. 5-{8-[(3,4'-Difluoro[1,1'-biphenyl]-4-yl)methoxy]-2-methylimidazo[1,2-a]pyridin-3-yl}-N-methylpyridine-2-carboxamide (1o) showed high in vitro potency and brain exposure, induced a robust reduction in brain Aß42 levels, and exhibited undetectable inhibition of cytochrome p450 enzymes. Moreover, compound 1o showed excellent efficacy against cognitive deficits in AD model mice. These findings suggest that compound 1o is a promising candidate for AD therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Cognitive Dysfunction/drug therapy , Disease Models, Animal , Imidazoles/pharmacology , Pyridines/pharmacology , Administration, Oral , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Line, Tumor , Cognitive Dysfunction/metabolism , Dose-Response Relationship, Drug , Humans , Imidazoles/administration & dosage , Imidazoles/chemistry , Male , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
2.
Bioorg Med Chem ; 28(1): 115132, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31767402

ABSTRACT

Gamma-secretase modulators (GSMs) are promising disease-modifying drugs for Alzheimer's disease because they can selectively decrease pathogenic amyloid-ß42 (Aß42) levels. Here we report the discovery of orally active N-ethylpyridine-2-carboxamide derivatives as GSMs. The isoindolinone moiety of 5-[8-(benzyloxy)-2-methylimidazo[1,2-a]pyridin-3-yl]-2-ethyl-2,3-dihydro-1H-isoindol-1-one hydrogen chloride (1a) was replaced with a picolinamide moiety. Optimization of the benzyl group significantly improved GSM activity and mouse microsomal stability. 5-{8-[([1,1'-Biphenyl]-4-yl)methoxy]-2-methylimidazo[1,2-a]pyridin-3-yl}-N-ethylpyridine-2-carboxamide hydrogen chloride (1v) potently reduced Aß42 levels with an IC50 value of 0.091 µM in cultured cells without inhibiting CYP3A4. Moreover, 1v demonstrated a sustained pharmacokinetic profile and significantly reduced brain Aß42 levels in mice.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Drug Discovery , Enzyme Inhibitors/pharmacology , Pyridines/pharmacology , Administration, Oral , Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Male , Mice , Mice, Inbred Strains , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Structure-Activity Relationship
3.
Bioorg Med Chem ; 26(2): 435-442, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29249626

ABSTRACT

Gamma-secretase modulators (GSMs) selectively inhibit the production of amyloid-ß 42 (Aß42) and may therefore be useful in the management of Alzheimer's disease. Most heterocyclic GSMs that are not derived from nonsteroidal anti-inflammatory drugs contain an arylimidazole moiety that potentially inhibits cytochrome P450 (CYP) activity. Here, we discovered imidazopyridine derivatives that represent a new class of scaffold for GSMs, which do not have a strongly basic end group such as arylimidazole. High-throughput screening identified 2-methyl-8-[(2-methylbenzyl)oxy]-3-(pyridin-4-yl)imidazo[1,2-a]pyridine (3a), which inhibited the cellular production of Aß42 (IC50 = 7.1 µM) without changing total production of Aß. Structural optimization of this series of compounds identified 5-[8-(benzyloxy)-2-methylimidazo[1,2-a]pyridin-3-yl]-2-ethylisoindolin-1-one (3m) as a potent inhibitor of Aß42 (IC50 = 0.39 µM) but not CYP3A4. Further, 3m demonstrated a sustained pharmacokinetic profile in mice and sufficiently penetrated the brain.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Drug Discovery , Heterocyclic Compounds/pharmacology , Imidazoles/pharmacology , Pyridines/pharmacology , Administration, Oral , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Animals , Cell Line, Tumor , Cytochrome P-450 CYP3A/metabolism , Dose-Response Relationship, Drug , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/chemistry , Humans , Imidazoles/administration & dosage , Imidazoles/chemistry , Injections, Intraperitoneal , Male , Mice , Mice, Inbred Strains , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/biosynthesis , Pyridines/administration & dosage , Pyridines/chemistry , Structure-Activity Relationship
4.
Brain Res ; 1605: 49-58, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25698614

ABSTRACT

Stem cell transplantation therapy is currently in clinical trials for the treatment of ischemic stroke, and several beneficial aspects have been reported. Similarly, in Alzheimer's disease (AD), stem cell therapy is expected to provide an efficient therapeutic approach. Indeed, the intracerebral transplantation of stem cells reduced amyloid-ß (Aß) deposition and rescued memory deficits in AD model mice. Here, we show that intravenous transplantation of bone marrow-derived mononuclear cells (BMMCs) improves cognitive function in two different AD mouse models, DAL and APP mice, and prevents neurodegeneration. GFP-positive BMMCs were isolated from tibiae and femurs of 4-week-old mice and then transplanted intravenously into DAL and APP mice. Transplantation of BMMCs suppressed neuronal loss and restored memory impairment of DAL mice to almost the same level as in wild-type mice. Transplantation of BMMCs to APP mice reduced Aß deposition in the brain. APP mice treated with BMMCs performed significantly better on behavioral tests than vehicle-injected mice. Moreover, the effects were observed even with transplantation after the onset of cognitive impairment in DAL mice. Together, our results indicate that intravenous transplantation of BMMCs has preventive effects against the cognitive decline in AD model mice and suggest a potential therapeutic effect of BMMC transplantation therapy.


Subject(s)
Alzheimer Disease/therapy , Bone Marrow Transplantation/methods , Cognition Disorders/prevention & control , Leukocytes, Mononuclear/transplantation , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Bone Marrow Cells , Cognition Disorders/etiology , Disease Models, Animal , Female , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Plaque, Amyloid/metabolism
5.
Neurosci Lett ; 587: 126-31, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25529196

ABSTRACT

Oxidative stress is known to play a prominent role in the onset and early stage progression of Alzheimer's disease (AD). For example, protein oxidation and lipid peroxidation levels are increased in patients with mild cognitive impairment. Here, we created a double-transgenic mouse model of AD to explore the pathological and behavioral effects of oxidative stress. Double transgenic (APP/DAL) mice were constructed by crossing Tg2576 (APP) mice, which express a mutant form of human amyloid precursor protein (APP), with DAL mice expressing a dominant-negative mutant of mitochondrial aldehyde dehydrogenase 2 (ALDH2), in which oxidative stress is enhanced. Y-maze and object recognition tests were performed at 3 and 6 months of age to evaluate learning and memory. The accumulation of amyloid plaques, deposition of phosphorylated-tau protein, and number of astrocytes in the brain were assessed histopathologically at 3, 6, 9, and 12-15 months of age. The life span of APP/DAL mice was significantly shorter than that of APP or DAL mice. In addition, they showed accelerated amyloid deposition, tau phosphorylation, and gliosis. Furthermore, these mice showed impaired performance on Y-maze and object recognition tests at 3 months of age. These data suggest that oxidative stress accelerates cognitive dysfunction and pathological insults in the brain. APP/DAL mice could be a useful model for exploring new approaches to AD treatment.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid/metabolism , Memory Disorders/psychology , Oxidative Stress , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Gliosis , Learning , Memory Disorders/genetics , Mice, Transgenic , Mitochondrial Proteins/genetics , Phosphorylation , tau Proteins/metabolism
6.
Neuropharmacology ; 79: 412-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24373902

ABSTRACT

γ-Secretase is the enzyme responsible for the intramembranous proteolysis of various substrates, such as amyloid precursor protein (APP) and Notch. Amyloid-ß peptide 42 (Aß42) is produced through the sequential proteolytic cleavage of APP by ß- and γ-secretase and causes the synaptic dysfunction associated with memory impairment in Alzheimer's disease. Here, we identified a novel cyclohexylamine-derived γ-secretase modulator, {(1R*,2S*,3R*)-3-[(cyclohexylmethyl)(3,3-dimethylbutyl)amino]-2-[4-(trifluoromethyl)phenyl]cyclohexyl}acetic acid (AS2715348), that may inhibit this pathological response. AS2715348 was seen to reduce both cell-free and cellular production of Aß42 without increasing levels of APP ß-carboxyl terminal fragment or inhibiting Notch signaling. Additionally, the compound increased Aß38 production, suggesting a shift of the cleavage site in APP. The inhibitory potency of AS2715348 on endogenous Aß42 production was similar across human, mouse, and rat cells. Oral administration with AS2715348 at 1 mg/kg and greater significantly reduced brain Aß42 levels in rats, and no Notch-related toxicity was observed after 28-day treatment at 100 mg/kg. Further, AS2715348 significantly ameliorated cognitive deficits in APP-transgenic Tg2576 mice. Finally, AS2715348 significantly reduced brain Aß42 levels in cynomolgus monkeys. These findings collectively show the promise for AS2715348 as a potential disease-modifying drug for Alzheimer's disease.


Subject(s)
Acetates/pharmacology , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Brain/drug effects , Cyclohexylamines/pharmacology , Neuroprotective Agents/pharmacology , Acetates/adverse effects , Acetates/pharmacokinetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Cell Line, Tumor , Cognition/drug effects , Cyclohexylamines/adverse effects , Cyclohexylamines/pharmacokinetics , Disease Models, Animal , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Transgenic , Molecular Structure , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Nootropic Agents/adverse effects , Nootropic Agents/chemistry , Nootropic Agents/pharmacology , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Notch/metabolism
7.
J Neurochem ; 125(3): 465-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23240999

ABSTRACT

Given that amyloid-ß 42 (Aß42) is believed to be a culprit in Alzheimer's disease (AD), reducing Aß42 production should be a potential therapeutic approach. γ-Secretase modulators (GSMs) cause selective reduction of Aß42 or both reduction of Aß42 and Aß40 without affecting total Aß through shifting the γ-cleavage position in amyloid precursor protein. We recently reported on GSM-2, one of the second-generation GSMs, that selectively reduced brain Aß42 level and significantly ameliorated cognitive deficits in plaque-free 5.5-month-old Tg2576 AD model mice. Here, we investigated the effects of GSM-2 on 10-, 14-, and 18-month-old mice which had age-dependent increase in amyloid plaques. Eight-day treatment with GSM-2 significantly ameliorated cognitive deficits measured by Y-maze task in the mice of any age. However, GSM-2 reduced brain soluble Aß42 only in 10-month-old mice. In contrast, GSM-2 markedly reduced newly synthesized soluble Aß42 in both 10- and 18-month-old mice with similar efficacy when measured using the stable isotope-labeling technique, suggesting that nascent Aß42 plays a more significant role than plaque-associated soluble Aß42 in the cognitive deterioration of Tg2576 mice. These findings further indicate the potential utility of approach to reducing Aß42 synthesis in AD therapeutic regimens.


Subject(s)
Alzheimer Disease/complications , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Cognition Disorders/metabolism , Enzyme Inhibitors/therapeutic use , Peptide Fragments/metabolism , Acetates/pharmacology , Acetates/therapeutic use , Age Factors , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/therapeutic use , Chromatography, Liquid , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Enzyme Inhibitors/chemistry , Enzyme-Linked Immunosorbent Assay , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mass Spectrometry , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peptide Fragments/immunology , Piperidines/pharmacology , Piperidines/therapeutic use
8.
J Neurosci ; 32(6): 2037-50, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22323718

ABSTRACT

γ-Secretase inhibitors (GSIs) reduce amyloid-ß (Aß) peptides but inevitably increase the ß-C-terminal fragment (ß-CTF) of amyloid precursor protein (APP), potentially having undesirable effects on synapses. In contrast, γ-secretase modulators (GSMs) reduce Aß42 without increasing ß-CTF. Although the Aß-lowering effects of these compounds have been extensively studied, little effort has been made to investigate their effects on cognition. Here, we compared the effects of two GSIs--(2S)-2-hydroxy-3-methyl-N-[(2S)-1-{[(1S)-3-methyl-2-oxo-2,3,4,5-tetrahydro-1H-3-benzazepin-1-yl]amino}-1-oxopropan-2-yl]butanamide (LY450139, semagacestat) and (2R)-2-[[(4-chlorophenyl)sulfonyl][[2-fluoro-4-(1,2,4-oxazol-3-yl)phenyl]methyl]amino-5,5,5-trifluoropentanamide (BMS-708163)--and a second-generation GSM [{(2S,4R)-1-[(4R)-1,1,1-trifluoro-7-methyloctan-4-yl]-2-[4-(trifluoromethyl)phenyl]piperidin-4-yl}acetic acid (GSM-2)] on spatial working memory in APP-transgenic (Tg2576) and nontransgenic mice using the Y-maze task. While acute dosing with either GSI ameliorated memory deficits in 5.5-month-old Tg2576 mice, these effects disappeared after 8 d subchronic dosing. Subchronic dosing with either GSI rather impaired normal cognition in 3-month-old Tg2576 mice, with no inhibition on the processing of other γ-secretase substrates, such as Notch, N-cadherin, or EphA4, in the brain. LY450139 also impaired normal cognition in wild-type mice; however, the potency was 10-fold lower than that in Tg2576 mice, indicating an APP-dependent mechanism likely with ß-CTF accumulation. Immunofluorescence studies revealed that the ß-CTF accumulation was localized in the presynaptic terminals of the hippocampal stratum lucidum and dentate hilus, implying an effect on presynaptic function in the mossy fibers. In contrast, both acute and subchronic dosing with GSM-2 significantly ameliorated memory deficits in Tg2576 mice and did not affect normal cognition in wild-type mice. We demonstrated a clear difference between GSI and GSM in effects on functional consequences, providing new insights into strategies for developing these drugs against Alzheimer's disease.


Subject(s)
Alanine/analogs & derivatives , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/physiology , Azepines/pharmacology , Cognition/drug effects , Cognition/physiology , Protease Inhibitors/pharmacology , Alanine/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Tumor , Female , Humans , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic
9.
J Biol Chem ; 283(10): 6594-606, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18182390

ABSTRACT

Recent evidence suggests that unscheduled cell cycle activity leads to neuronal cell death. 3-Nitropropionic acid (3-NP) is an irreversible inhibitor of succinate dehydrogenase and induces cell death in both striatum and cerebral cortex. Here we analyzed the involvement of aberrant cell cycle progression in 3-NP-induced cell death in these brain regions. 3-NP reduced the level of cyclin-dependent kinase inhibitor p27 in striatum but not in cerebral cortex. 3-NP also induced phosphorylation of retinoblastoma protein, a marker of cell cycle progression at late G(1) phase, only in striatum. Pharmacological experiments revealed that cyclin-dependent kinase activity and N-methyl-d-aspartate (NMDA) receptor were cooperatively involved in cell death by 3-NP in striatal neurons, whereas only NMDA receptor was involved in 3-NP-induced neurotoxicity in cortical neurons. Death of striatal neurons was preceded by elevation of somatic Ca(2+) and activation of calpain, a Ca(2+)-dependent protease. Both striatal p27 down-regulation and cell death provoked by 3-NP were dependent on calpain activity. Moreover, transfection of p27 small interfering RNA reduced striatal cell viability. In cortical neurons, however, there was no change in somatic Ca(2+) and calpain activity by 3-NP, and calpain inhibitors were not protective. These results suggest that 3-NP induces aberrant cell cycle progression and neuronal cell death via p27 down-regulation by calpain in striatum but not in the cerebral cortex. This is the first report for differential involvement of cell cycle reactivation in different brain regions and lightens the mechanism for region-selective vulnerability in human disease, including Huntington disease.


Subject(s)
Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Enzyme Inhibitors/toxicity , G1 Phase/drug effects , Nitro Compounds/toxicity , Propionates/toxicity , Succinate Dehydrogenase/antagonists & inhibitors , Animals , Calcium Signaling/drug effects , Calpain/metabolism , Cell Death/drug effects , Cerebral Cortex/pathology , Convulsants/toxicity , Corpus Striatum/pathology , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , Organ Specificity/drug effects , Phosphorylation/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/biosynthesis , Retinoblastoma Protein/metabolism , Succinate Dehydrogenase/metabolism
10.
Neuroreport ; 18(17): 1781-5, 2007 Nov 19.
Article in English | MEDLINE | ID: mdl-18090311

ABSTRACT

p27 is a cyclin-dependent kinase inhibitor which arrests cell cycle at G1-S phase. Using RNA interference method, we previously showed that reduction of endogenous p27 expression induces cell death through cell cycle progression in cultured cortical neurons. In this study, we investigated responses to kainate treatment using p27 knockout mice. Injection of kainic acid induced p27 downregulation and retinoblastoma protein phosphorylation in wild-type mouse hippocampus. No change was observed in hippocampal cell viability in untreated adult p27 heterozygous and homozygous mice compared with wild type (+/+). p27 homozygous mice, however, displayed enhanced seizure and hippocampal degeneration after kainic acid treatment. This study first suggests that ablation of p27 enhance kainate-induced seizure and hippocampal cell death in vivo.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/physiology , Excitatory Amino Acid Agonists/toxicity , Hippocampus/pathology , Kainic Acid/antagonists & inhibitors , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Seizures/chemically induced , Seizures/physiopathology , Animals , Blotting, Western , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor p27/genetics , DNA/genetics , Immunohistochemistry , Kainic Acid/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Seizures/genetics
11.
J Neurochem ; 99(3): 733-44, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16824045

ABSTRACT

Recent evidence suggests that cell cycle-related molecules play pivotal roles in multiple forms of cell death in post-mitotic neurons. Nevertheless, it remains unclear what molecular mechanisms are involved in the regulation of expression levels and activities of these molecules. We showed previously that treatment with extracellular glutamate decreases cyclin-dependent kinase inhibitor p27 before neuronal cell death. In this study, we demonstrate that reductions of both p27 and neuronal viability were dependent on activity of calpain, a Ca(2+)-dependent protease, but not on activity of caspase 3. Interestingly, the glutamate-induced reduction of p27 was not dependent on the ubiquitin-proteasome system. In fact, p27 was present only in the neuronal nucleus, whereas calpain 1, a ubiquitous calpain, was observed both in the neuronal nucleus and cytoplasm in control cultures. Glutamate treatment did not change the localization patterns of p27 and calpain 1. It reduced p27 expression level in the nucleus in a calpain-dependent manner. In vitro experiments using neuronal cell lysate and p27 recombinant protein revealed that p27 was degraded as a substrate of activated calpain 1. These results suggest that calpain(s), activated by glutamate treatment, degrade(s) p27 in the nucleus of neurons, which might promote aberrant cell cycle progression.


Subject(s)
Calpain/metabolism , Cerebral Cortex/metabolism , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Glutamic Acid/pharmacology , Neurons/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Calpain/pharmacology , Caspase 3/metabolism , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Cytoplasm/drug effects , Cytoplasm/enzymology , Cytoplasm/metabolism , Down-Regulation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Immunohistochemistry , Neurons/drug effects , Neurons/ultrastructure , Proteasome Endopeptidase Complex/metabolism , Rats , Signal Transduction/drug effects , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...