Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Proc Natl Acad Sci U S A ; 112(49): 15220-5, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26598704

ABSTRACT

Therapeutic options for the restoration of neurological functions after acute axonal injury are severely limited. In addition to limiting neuronal loss, effective treatments face the challenge of restoring axonal growth within an injury environment where inhibitory molecules from damaged myelin and activated astrocytes act as molecular and physical barriers. Overcoming these barriers to permit axon growth is critical for the development of any repair strategy in the central nervous system. Here, we identify poly(ADP-ribose) polymerase 1 (PARP1) as a previously unidentified and critical mediator of multiple growth-inhibitory signals. We show that exposure of neurons to growth-limiting molecules--such as myelin-derived Nogo and myelin-associated glycoprotein--or reactive astrocyte-produced chondroitin sulfate proteoglycans activates PARP1, resulting in the accumulation of poly(ADP-ribose) in the cell body and axon and limited axonal growth. Accordingly, we find that pharmacological inhibition or genetic loss of PARP1 markedly facilitates axon regeneration over nonpermissive substrates. Together, our findings provide critical insights into the molecular mechanisms of axon growth inhibition and identify PARP1 as an effective target to promote axon regeneration.


Subject(s)
Axons , Enzyme Inhibitors/pharmacology , Nerve Regeneration , Poly(ADP-ribose) Polymerases/metabolism , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/drug effects
2.
J Neurosci ; 33(20): 8621-32, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23678107

ABSTRACT

Histone deacetylase (HDAC) inhibitors have been used to promote neuronal survival and ameliorate neurological dysfunction in a host of neurodegenerative disease models. The precise molecular mechanisms whereby HDAC inhibitors prevent neuronal death are currently the focus of intensive research. Here we demonstrate that HDAC inhibition prevents DNA damage-induced neurodegeneration by modifying the acetylation pattern of the tumor suppressor p53, which decreases its DNA-binding and transcriptional activation of target genes. Specifically, we identify that acetylation at K382 and K381 prevents p53 from associating with the pro-apoptotic PUMA gene promoter, activating transcription, and inducing apoptosis in mouse primary cortical neurons. Paradoxically, acetylation of p53 at the same lysines in various cancer cell lines leads to the induction of PUMA expression and death. Together, our data provide a molecular understanding of the specific outcomes of HDAC inhibition and suggest that strategies aimed at enhancing p53 acetylation at K381 and K382 might be therapeutically viable for capturing the beneficial effects in the CNS, without compromising tumor suppression.


Subject(s)
Apoptosis/physiology , DNA Damage/physiology , Histone Deacetylases/metabolism , Neurons/physiology , Tumor Suppressor Protein p53/metabolism , Acetylation , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Brain/cytology , Cells, Cultured , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/drug effects , DNA Damage/genetics , Electroporation , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Histone Deacetylases/genetics , Humans , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Microarray Analysis , Mutagenesis, Site-Directed/methods , Mutation/genetics , Neurons/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism
3.
Neurobiol Dis ; 49: 13-21, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22944173

ABSTRACT

Preventing neuronal death is a priority for treating neurological diseases. However, therapies that inhibit pathological neuron loss could promote tumorigenesis by preventing the physiological death of cancerous cells. To avert this, we targeted the transcriptional upregulation of p21(waf1/cip1) (p21), an endogenous tumor suppressor with neuroprotective and pro-regenerative activity. We identified potential p21 indcuers by screening a FDA-approved drug and natural product small molecule library against hippocampal HT22 cells stably expressing a luciferase reporter driven by the proximal 60bp of the p21 promoter, and tested them for neuroprotection from glutathione depletion mediated oxidative stress, and cytotoxicity to cancer cell lines (DLD-1, Neuro-2A, SH-SY5Y, NGP, CHLA15, CHP212, and SK-N-SH) in vitro. Of the p21 inducers identified, only ciclopirox, a hypoxia-inducible factor prolyl-4-hydroxylase (HIF-PHD) inhibitor, simultaneously protected neurons from glutathione depletion and decreased cancer cell proliferation at concentrations that were not basally toxic to neurons. We found that other structurally distinct HIF-PHD inhibitors (desferrioxamine, 3,4-dihydroxybenzoate, and dimethyloxalyl glycine) also protected neurons at concentrations that killed cancer cells. HIF-PHD inhibitors stabilize HIF transcription factors, mediating genetic adaptation to hypoxia. While augmenting HIF stability is believed to promote tumorigenesis, we found that chronic HIF-PHD inhibition killed cancer cells, suggesting a protumorigenic role for these enzymes. Moreover, our findings suggest that PHD inhibitors can be used to treat neurological disease without significant concern for cell-autonomous tumor promotion.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Neuroprotective Agents/pharmacology , Prolyl-Hydroxylase Inhibitors/pharmacology , Animals , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cells, Cultured , Drug Screening Assays, Antitumor , Hippocampus/drug effects , Hippocampus/physiology , Humans , Mice , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology , PC12 Cells , Rats , Small Molecule Libraries
4.
Proc Natl Acad Sci U S A ; 107(40): 17385-90, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20855618

ABSTRACT

Neurons rely on their metabolic coupling with astrocytes to combat oxidative stress. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) appears important for astrocyte-dependent neuroprotection from oxidative insults. Indeed, Nrf2 activators are effective in stroke, Parkinson disease, and Huntington disease models. However, key endogenous signals that initiate adaptive neuroprotective cascades in astrocytes, including activation of Nrf2-mediated gene expression, remain unclear. Hydrogen peroxide (H(2)O(2)) plays an important role in cell signaling and is an attractive candidate mediator of adaptive responses in astrocytes. Here we determine (i) the significance of H(2)O(2) in promoting astrocyte-dependent neuroprotection from oxidative stress, and (ii) the relevance of H(2)O(2) in inducing astrocytic Nrf2 activation. To control the duration and level of cytoplasmic H(2)O(2) production in astrocytes cocultured with neurons, we heterologously expressed the H(2)O(2)-producing enzyme Rhodotorula gracilis D-amino acid oxidase (rgDAAO) selectively in astrocytes. Exposure of rgDAAO-astrocytes to D-alanine lead to the concentration-dependent generation of H(2)O(2). Seven hours of low-level H(2)O(2) production (Ć¢ĀˆĀ¼3.7 nmolĀ·minĀ·mg protein) in astrocytes protected neurons from oxidative stress, but higher levels (Ć¢ĀˆĀ¼130 nmolĀ·minĀ·mg protein) were neurotoxic. Neuroprotection occurred without direct neuronal exposure to astrocyte-derived H(2)O(2), suggesting a mechanism specific to astrocytic intracellular signaling. Nrf2 activation mimicked the effect of astrocytic H(2)O(2) yet H(2)O(2)-induced protection was independent of Nrf2. Astrocytic protein tyrosine phosphatase inhibition also protected neurons from oxidative death, representing a plausible mechanism for H(2)O(2)-induced neuroprotection. These findings demonstrate the utility of rgDAAO for spatially and temporally controlling intracellular H(2)O(2) concentrations to uncover unique astrocyte-dependent neuroprotective mechanisms.


Subject(s)
Astrocytes/metabolism , Hydrogen Peroxide/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Oxidants/metabolism , Oxidative Stress/physiology , Animals , Astrocytes/cytology , Cells, Cultured , Coculture Techniques , D-Amino-Acid Oxidase/metabolism , Glutathione/metabolism , Microarray Analysis , NF-E2-Related Factor 2/metabolism , Neurons/cytology , Rats , Rhodotorula/enzymology
5.
Exp Neurol ; 368: 114479, 2023 10.
Article in English | MEDLINE | ID: mdl-37454712

ABSTRACT

Spinal cord injury (SCI)-induced tissue damage spreads to neighboring spared cells in the hours, days, and weeks following injury, leading to exacerbation of tissue damage and functional deficits. Among the biochemical changes is the rapid reduction of cellular nicotinamide adenine dinucleotide (NAD+), an essential coenzyme for energy metabolism and an essential cofactor for non-redox NAD+-dependent enzymes with critical functions in sensing and repairing damaged tissue. NAD+ depletion propagates tissue damage. Augmenting NAD+ by exogenous application of NAD+, its synthesizing enzymes, or its cellular precursors mitigates tissue damage. Nicotinamide riboside (NR) is considered to be one of the most promising NAD+ precursors for clinical application due to its ability to safely and effectively boost cellular NAD+ synthesis in rats and humans. Moreover, various preclinical studies have demonstrated that NR can provide tissue protection. Despite these promising findings, little is known about the potential benefits of NR in the context of SCI. In the current study, we tested whether NR administration could effectively increase NAD+ levels in the injured spinal cord and whether this augmentation of NAD+ would promote spinal cord tissue protection and ultimately lead to improvements in locomotor function. Our findings indicate that administering NR (500Ā mg/kg) intraperitoneally from four days before to two weeks after a mid-thoracic contusion-SCI injury, effectively doubles NAD+ levels in the spinal cord of Long-Evans rats. Moreover, NR administration plays a protective role in preserving spinal cord tissue post-injury, particularly in neurons and axons, as evident from the observed gray and white matter sparing. Additionally, it enhances motor function, as evaluated through the BBB subscore and missteps on the horizontal ladderwalk. Collectively, these findings demonstrate that administering NR, a precursor of NAD+, increases NAD+ within the injured spinal cord and effectively mitigates the tissue damage and functional decline that occurs following SCI.


Subject(s)
NAD , Spinal Cord Injuries , Humans , Rats , Animals , NAD/metabolism , Rats, Long-Evans , Niacinamide/pharmacology , Niacinamide/therapeutic use , Niacinamide/metabolism , Pyridinium Compounds , Spinal Cord Injuries/drug therapy
6.
J Neurosci ; 31(18): 6858-70, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21543616

ABSTRACT

Oncogenic transformation of postmitotic neurons triggers cell death, but the identity of genes critical for degeneration remain unclear. The antitumor antibiotic mithramycin prolongs survival of mouse models of Huntington's disease in vivo and inhibits oxidative stress-induced death in cortical neurons in vitro. We had correlated protection by mithramycin with its ability to bind to GC-rich DNA and globally displace Sp1 family transcription factors. To understand how antitumor drugs prevent neurodegeneration, here we use structure-activity relationships of mithramycin analogs to discover that selective DNA-binding inhibition of the drug is necessary for its neuroprotective effect. We identify several genes (Myc, c-Src, Hif1α, and p21(waf1/cip1)) involved in neoplastic transformation, whose altered expression correlates with protective doses of mithramycin or its analogs. Most interestingly, inhibition of one these genes, Myc, is neuroprotective, whereas forced expression of Myc induces Rattus norvegicus neuronal cell death. These results support a model in which cancer cell transformation shares key genetic components with neurodegeneration.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Neurons/drug effects , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Sp1 Transcription Factor/metabolism , Analysis of Variance , Animals , Animals, Genetically Modified , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chromatin Immunoprecipitation , Drosophila , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Sp1 Transcription Factor/genetics , Structure-Activity Relationship
7.
Proc Natl Acad Sci U S A ; 106(46): 19599-604, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19884510

ABSTRACT

Central nervous system (CNS) trauma can result in tissue disruption, neuronal and axonal degeneration, and neurological dysfunction. The limited spontaneous CNS repair in adulthood and aging is often insufficient to overcome disability. Several investigations have demonstrated that targeting HDAC activity can protect neurons and glia and improve outcomes in CNS injury and disease models. However, the enthusiasm for pan-HDAC inhibition in treating neurological conditions is tempered by their toxicity toward a host of CNS cell types -a biological extension of their anticancer properties. Identification of the HDAC isoform, or isoforms, that specifically mediate the beneficial effects of pan-HDAC inhibition could overcome this concern. Here, we show that pan-HDAC inhibition not only promotes neuronal protection against oxidative stress, a common mediator of injury in many neurological conditions, but also promotes neurite growth on myelin-associated glycoprotein and chondroitin sulfate proteoglycan substrates. Real-time PCR revealed a robust and selective increase in HDAC6 expression due to injury in neurons. Accordingly, we have used pharmacological and genetic approaches to demonstrate that inhibition of HDAC6 can promote survival and regeneration of neurons. Consistent with a cytoplasmic localization, the biological effects of HDAC6 inhibition appear transcription-independent. Notably, we find that selective inhibition of HDAC6 avoids cell death associated with pan-HDAC inhibition. Together, these findings define HDAC6 as a potential nontoxic therapeutic target for ameliorating CNS injury characterized by oxidative stress-induced neurodegeneration and insufficient axonal regeneration.


Subject(s)
Central Nervous System/injuries , Central Nervous System/physiology , Histone Deacetylases/metabolism , Nerve Regeneration , Neurites/physiology , Neurons/physiology , Animals , Apoptosis , Central Nervous System/enzymology , Cerebral Cortex/enzymology , Cerebral Cortex/physiology , Ganglia, Spinal/enzymology , Ganglia, Spinal/physiology , Histone Deacetylase 6 , Histone Deacetylases/genetics , Male , Neurites/enzymology , Neurodegenerative Diseases/enzymology , Neurons/enzymology , Oxidative Stress , RNA Interference , Rats , Rats, Sprague-Dawley
8.
J Med Chem ; 64(8): 4810-4840, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33830764

ABSTRACT

Histone deacetylase 6 (HDAC6) is a promising therapeutic target for the treatment of neurodegenerative disorders. SW-100 (1a), a phenylhydroxamate-based HDAC6 inhibitor (HDAC6i) bearing a tetrahydroquinoline (THQ) capping group, is a highly potent and selective HDAC6i that was shown to be effective in mouse models of Fragile X syndrome and Charcot-Marie-Tooth disease type 2A (CMT2A). In this study, we report the discovery of a new THQ-capped HDAC6i, termed SW-101 (1s), that possesses excellent HDAC6 potency and selectivity, together with markedly improved metabolic stability and druglike properties compared to SW-100 (1a). X-ray crystallography data reveal the molecular basis of HDAC6 inhibition by SW-101 (1s). Importantly, we demonstrate that SW-101 (1s) treatment elevates the impaired level of acetylated α-tubulin in the distal sciatic nerve, counteracts progressive motor dysfunction, and ameliorates neuropathic symptoms in a CMT2A mouse model bearing mutant MFN2. Taken together, these results bode well for the further development of SW-101 (1s) as a disease-modifying HDAC6i.


Subject(s)
Charcot-Marie-Tooth Disease/drug therapy , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Quinolines/chemistry , Acetylation , Animals , Benzamides/chemistry , Benzamides/metabolism , Binding Sites , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Crystallography, X-Ray , Disease Models, Animal , Half-Life , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Phenotype , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Quinolines/metabolism , Quinolines/therapeutic use , Structure-Activity Relationship , Tubulin/metabolism
9.
J Am Chem Soc ; 132(31): 10842-6, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20614936

ABSTRACT

Structure-based drug design combined with homology modeling techniques were used to develop potent inhibitors of HDAC6 that display superior selectivity for the HDAC6 isozyme compared to other inhibitors. These inhibitors can be assembled in a few synthetic steps, and thus are readily scaled up for in vivo studies. An optimized compound from this series, designated Tubastatin A, was tested in primary cortical neuron cultures in which it was found to induce elevated levels of acetylated alpha-tubulin, but not histone, consistent with its HDAC6 selectivity. Tubastatin A also conferred dose-dependent protection in primary cortical neuron cultures against glutathione depletion-induced oxidative stress. Importantly, when given alone at all concentrations tested, this hydroxamate-containing HDAC6-selective compound displayed no neuronal toxicity, thus, forecasting the potential application of this agent and its analogues to neurodegenerative conditions.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Anilides/chemistry , Anilides/pharmacology , Cell Death/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone Deacetylase 6 , Homocysteine/analogs & derivatives , Homocysteine/pharmacology , Humans , Hydroxamic Acids/chemistry , Indoles/chemistry , Models, Molecular , Molecular Structure , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Oxidative Stress/drug effects , Stereoisomerism , Structure-Activity Relationship
10.
Biochem Biophys Res Commun ; 393(4): 673-7, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20170631

ABSTRACT

Most olfactory bulb (OB) interneurons are derived from neural stem cells in the subventricular zone (SVZ) and migrate to the OB via the rostral migratory stream (RMS). Mature dopaminergic interneurons in the OB glomerular layer are readily identified by their synaptic activity-dependent expression of tyrosine hydroxylase (TH). Paradoxically, TH is not expressed in neural progenitors migrating in the RMS, even though ambient GABA and glutamate depolarize these progenitors. In forebrain slice cultures prepared from transgenic mice containing a GFP reporter gene under the control of the Th 9kb upstream regulatory region, treatment with histone deacetylase (HDAC) inhibitors (either sodium butyrate, Trichostatin A or Scriptaid) induced Th-GFP expression specifically in the RMS independently of depolarizing conditions in the culture media. Th-GFP expression in the glomerular layer was also increased in slices treated with Trichostatin A, but this increased expression was dependent on depolarizing concentrations of KCl in the culture media. Th-GFP expression was also induced in the RMS in vivo by intra-peritoneal injections with either sodium butyrate or valproic acid. Quantitative RT-PCR analysis of neurosphere cultures confirmed that HDAC inhibitors de-repressed Th expression in SVZ-derived neural progenitors. Together, these findings suggest that HDAC function is critical for regulating Th expression levels in both neural progenitors and mature OB dopaminergic neurons. However, the differential responses to the combinatorial exposure of HDAC inhibitors and depolarizing culture conditions indicate that Th expression in mature OB neurons and neural progenitors in the RMS are regulated by distinct HDAC-mediated mechanisms.


Subject(s)
Cell Movement , Histone Deacetylases/metabolism , Neurons/physiology , Olfactory Bulb/drug effects , Stem Cells/physiology , Tyrosine 3-Monooxygenase/biosynthesis , Animals , Butyrates/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Hydroxylamines/pharmacology , Mice , Mice, Transgenic , Neurons/cytology , Neurons/enzymology , Olfactory Bulb/enzymology , Prosencephalon/cytology , Prosencephalon/drug effects , Prosencephalon/enzymology , Quinolines/pharmacology , Stem Cells/cytology , Stem Cells/enzymology , Tyrosine 3-Monooxygenase/genetics
11.
Exp Neurol ; 328: 113281, 2020 06.
Article in English | MEDLINE | ID: mdl-32147437

ABSTRACT

Charcot-Marie-Tooth type 2A (CMT2A) peripheral neuropathy, the most common axonal form of CMT, is caused by dominantly inherited point mutations in the Mitofusin 2 (Mfn2) gene. It is characterized by progressive length-dependent degeneration of motor and sensory nerves with corresponding clinical features of motor and sensory impairment. There is no cure for CMT, and therapeutic approaches are limited to physical therapy, orthopedic devices, surgery, and analgesics. In this study we focus on histone deacetylase 6 (HDAC6) as a therapeutic target in a mouse model of mutant MFN2 (MFN2R94Q)-induced CMT2A. We report that these mice display progressive motor and sensory dysfunction as well as a significant decrease in α-tubulin acetylation in distal segments of long peripheral nerves. Treatment with a new, highly selective HDAC6 inhibitor, SW-100, was able to restore α-tubulin acetylation and ameliorate motor and sensory dysfunction when given either prior to or after the onset of symptoms. To confirm HDAC6 is the target for ameliorating the CMT2A phenotype, we show that genetic deletion of Hdac6 in CMT2A mice prevents the development of motor and sensory dysfunction. Our findings suggest α-tubulin acetylation defects in distal parts of nerves as a pathogenic mechanism and HDAC6 as a therapeutic target for CMT2A.


Subject(s)
Benzamides/pharmacology , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Quinolines/pharmacology , Tubulin/metabolism , Acetylation/drug effects , Animals , Charcot-Marie-Tooth Disease/metabolism , Mice , Mice, Mutant Strains , Motor Activity/drug effects
12.
J Neurosci ; 28(1): 163-76, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18171934

ABSTRACT

Histone deacetylase (HDAC) inhibitors are currently in human clinical trials as antitumor drugs because of their ability to induce cell dysfunction and death in cancer cells. The toxic effects of HDAC inhibitors are also apparent in cortical neurons in vitro, despite the ability of these agents to induce significant protection in the cells they do not kill. Here we demonstrate that pulse exposure of cortical neurons (2 h) in an in vitro model of oxidative stress results in durable neuroprotection without toxicity. Protection was associated with transcriptional upregulation of the cell cycle inhibitor, p21(waf1/cip1), both in this model and in an in vivo model of permanent ischemia. Transgenic overexpression of p21(waf1/cip1) in neurons can mimic the protective effect of HDAC inhibitors against oxidative stress-induced toxicity, including death induced by glutathione depletion or peroxide addition. The protective effect of p21(waf1/cip1) in the context of oxidative stress appears to be unrelated to its ability to act in the nucleus to inhibit cell cycle progression. However, although p21(waf1/cip1) is sufficient for neuroprotection, it is not necessary for HDAC inhibitor neuroprotection, because these agents can completely protect neurons cultured from p21(waf1/cip1)-null mice. Together these findings demonstrate (1) that pulse inhibition of HDACs in cortical neurons can induce neuroprotection without apparent toxicity; (2) that p21(waf1/cip1) is sufficient but not necessary to mimic the protective effects of HDAC inhibition; and (3) that oxidative stress in this model induces neuronal cell death via cell cycle-independent pathways that can be inhibited by a cytosolic, noncanonical action of p21(waf1/cip1).


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Histone Deacetylases/metabolism , Neurons/physiology , Oxidative Stress/physiology , Analysis of Variance , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Cerebral Cortex/cytology , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/therapeutic use , Glutamic Acid/toxicity , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Histone Deacetylase Inhibitors , Infarction, Middle Cerebral Artery/drug therapy , Neurons/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Transfection/methods
13.
J Neurosci ; 28(46): 11753-9, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19005036

ABSTRACT

It is becoming increasingly clear that epigenetic modifications are critical factors in the regulation of gene expression. With regard to the nervous system, epigenetic alterations play a role in a diverse set of processes and have been implicated in a variety of disorders. Gaining a more complete understanding of the essential components and underlying mechanisms involved in epigenetic regulation could lead to novel treatments for a number of neurological and psychiatric conditions.


Subject(s)
Brain Diseases/genetics , Brain Diseases/metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Nervous System/metabolism , Animals , Brain Diseases/physiopathology , Chromatin/genetics , Chromatin/metabolism , DNA Methylation/genetics , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Humans
14.
Stroke ; 40(8): 2899-905, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19478231

ABSTRACT

Achieving therapeutic efficacy in ischemic stroke represents one of the biggest challenges in translational neurobiology. Despite extensive efforts, tissue plasminogen activator remains the only available intervention for enhancing functional recovery in humans once a stroke has occurred. To expand the repertoire of therapeutic options in stroke, one must consider and target its diverse pathophysiologies that trigger cell loss in a manner that also permits and enhances neuronal plasticity and repair. Several converging lines of inquiry suggest that histone deacetylase (HDAC) inhibition could be a strategy to achieve these goals. Here, we review evidence that targeting HDACs with low-molecular-weight inhibitors significantly decreases neuronal injury and improves functional outcome in multiple preclinical models of focal ischemia. These salutary effects emanate, in part, from modifications of chromatin and nonchromatin proteins that enhance adaptive gene expression or adaptive protein function. Together, the findings suggest that HDAC inhibition is a strategy capable of targeting diverse pathophysiologies of stroke with a wide therapeutic window.


Subject(s)
Drug Delivery Systems/methods , Histone Deacetylase Inhibitors , Stroke/enzymology , Animals , Enzyme Inhibitors/administration & dosage , Histone Deacetylases/metabolism , Humans , Stroke/drug therapy , Treatment Outcome
15.
J Cell Biol ; 218(6): 1871-1890, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31068376

ABSTRACT

Inhibition of histone deacetylase 6 (HDAC6) was shown to support axon growth on the nonpermissive substrates myelin-associated glycoprotein (MAG) and chondroitin sulfate proteoglycans (CSPGs). Though HDAC6 deacetylates α-tubulin, we find that another HDAC6 substrate contributes to this axon growth failure. HDAC6 is known to impact transport of mitochondria, and we show that mitochondria accumulate in distal axons after HDAC6 inhibition. Miro and Milton proteins link mitochondria to motor proteins for axon transport. Exposing neurons to MAG and CSPGs decreases acetylation of Miro1 on Lysine 105 (K105) and decreases axonal mitochondrial transport. HDAC6 inhibition increases acetylated Miro1 in axons, and acetyl-mimetic Miro1 K105Q prevents CSPG-dependent decreases in mitochondrial transport and axon growth. MAG- and CSPG-dependent deacetylation of Miro1 requires RhoA/ROCK activation and downstream intracellular Ca2+ increase, and Miro1 K105Q prevents the decrease in axonal mitochondria seen with activated RhoA and elevated Ca2+ These data point to HDAC6-dependent deacetylation of Miro1 as a mediator of axon growth inhibition through decreased mitochondrial transport.


Subject(s)
Histone Deacetylase 6/genetics , Mitochondria/metabolism , Neurons/metabolism , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics , Acetylation/drug effects , Animals , Axonal Transport/drug effects , Axonal Transport/genetics , Calcium/metabolism , Chondroitin Sulfate Proteoglycans/pharmacology , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation , Histone Deacetylase 6/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Myelin-Associated Glycoprotein/pharmacology , Neurons/cytology , Neurons/drug effects , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Signal Transduction , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
16.
J Mol Med (Berl) ; 85(12): 1331-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18043901

ABSTRACT

The brain demands oxygen and glucose to fulfill its roles as the master regulator of body functions as diverse as bladder control and creative thinking. Chemical and electrical transmission in the nervous system is rapidly disrupted in stroke as a result of hypoxia and hypoglycemia. Despite being highly evolved in its architecture, the human brain appears to utilize phylogenetically conserved homeostatic strategies to combat hypoxia and ischemia. Specifically, several converging lines of inquiry have demonstrated that the transcription factor hypoxia-inducible factor-1 (HIF1-1) mediates the activation of a large cassette of genes involved in adaptation to hypoxia in surviving neurons after stroke. Accordingly, pharmacological or molecular approaches that engage hypoxic adaptation at the point of one of its sensors (e.g., inhibition of HIF prolyl 4 hydroxylases) leads to profound sparing of brain tissue and enhanced recovery of function. In this review, we discuss the potential mechanisms that could subserve protective and restorative effects of augmenting hypoxic adaptation in the brain. The strategy appears to involve HIF-dependent and HIF-independent pathways and more than 70 genes and proteins activated transcriptionally and post-transcriptionally that can act at cellular, local, and system levels to compensate for oxygen insufficiency. The breadth and depth of this homeostatic program offers a hopeful alternative to the current pessimism towards stroke therapeutics.


Subject(s)
Brain/drug effects , Enzyme Inhibitors/pharmacology , Hypoxia, Brain/drug therapy , Neuroprotective Agents/pharmacology , Oxygen/metabolism , Stroke/drug therapy , Adaptation, Physiological , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Enzyme Inhibitors/therapeutic use , Humans , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Hypoxia, Brain/physiopathology , Hypoxia-Inducible Factor 1/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/metabolism , Signal Transduction/drug effects , Stroke/metabolism , Stroke/pathology , Stroke/physiopathology
17.
eNeuro ; 5(1)2018.
Article in English | MEDLINE | ID: mdl-29497702

ABSTRACT

Damage to the CNS results in neuronal and axonal degeneration, and subsequent neurological dysfunction. Endogenous repair in the CNS is impeded by inhibitory chemical and physical barriers, such as chondroitin sulfate proteoglycans (CSPGs) and myelin-associated glycoprotein (MAG), which prevent axon regeneration. Previously, it has been demonstrated that the inhibition of axonal histone deacetylase-6 (HDAC6) can promote microtubule α-tubulin acetylation and restore the growth of CSPGs- and MAG-inhibited axons. Since the acetylation of α-tubulin is regulated by two opposing enzymes, HDAC6 (deacetylation) and α-tubulin acetyltransferase-1 (αTAT1; acetylation), we have investigated the regulation of these enzymes downstream of a growth inhibitory signal. Our findings show that exposure of primary mouse cortical neurons to soluble CSPGs and MAG substrates cause an acute and RhoA-kinase-dependent reduction in α-tubulin acetylation and αTAT1 protein levels, without changes to either HDAC6 levels or HDAC6 activity. The CSPGs- and MAG-induced reduction in αTAT1 occurs primarily in the distal and middle regions of neurites and reconstitution of αTAT1, either by Rho-associated kinase (ROCK) inhibition or lentiviral-mediated αTAT1 overexpression, can restore neurite growth. Lastly, we demonstrate that CSPGs and MAG signaling decreases αTAT1 levels posttranscriptionally via a ROCK-dependent increase in αTAT1 protein turnover. Together, these findings define αTAT1 as a novel potential therapeutic target for ameliorating CNS injury characterized by growth inhibitory substrates that are prohibitive to axonal regeneration.


Subject(s)
Acetyltransferases/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Regeneration , Neurites/enzymology , Neuronal Outgrowth , Tubulin/metabolism , Animals , Down-Regulation , Female , Histone Deacetylase 6/metabolism , Mice , Microtubule Proteins/metabolism , Signal Transduction , rho-Associated Kinases/metabolism
18.
J Med Chem ; 50(13): 3054-61, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17539623

ABSTRACT

We compare the ability of two structurally different classes of epigenetic modulators, namely, histone deacetylase (HDAC) inhibitors containing either a hydroxamate or a mercaptoacetamide as the zinc binding group, to protect cortical neurons in culture from oxidative stress-induced death. This study reveals that some of the mercaptoacetamide-based HDAC inhibitors are fully protective, whereas the hydroxamates show toxicity at higher concentrations. Our present results appear to be consistent with the possibility that the mercaptoacetamide-based HDAC inhibitors interact with a different subset of the HDAC isozymes [less activity at HDAC1 and 2 correlates with less inhibitor toxicity], or alternatively, are interacting selectively with only the cytoplasmic HDACs that are crucial for protection from oxidative stress.


Subject(s)
Acetamides/chemical synthesis , Histone Deacetylase Inhibitors , Hydroxamic Acids/chemical synthesis , Neurons/drug effects , Neuroprotective Agents/chemical synthesis , Sulfhydryl Compounds/chemical synthesis , Acetamides/chemistry , Acetamides/pharmacology , Acetylation , Animals , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Epigenesis, Genetic , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Histones/metabolism , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology
19.
Neurosci Lett ; 625: 26-33, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27155457

ABSTRACT

Traumatic brain injury (TBI) contributes to nearly a third of all injury-related deaths in the United States. For survivors of TBI, depending on severity, patients can be left with devastating neurological disabilities that include impaired cognition or memory, movement, sensation, or emotional function. Despite the efforts to identify novel therapeutics, the only strategy to combat TBI is risk reduction (helmets, seatbelts, removal of fall hazards, etc.). Enormous heterogeneity exists within TBI, and it depends on the severity, the location, and whether the injury was focal or diffuse. Evidence from recent studies support the involvement of epigenetic mechanisms such as DNA methylation, chromatin post-translational modification, and miRNA regulation of gene expression in the post-injured brain. In this review, we discuss studies that have assessed epigenetic changes and mechanisms following TBI, how epigenetic changes might not only be limited to the nucleus but also impact the mitochondria, and the implications of these changes with regard to TBI recovery.


Subject(s)
Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/therapy , Epigenesis, Genetic , Animals , DNA Methylation , DNA, Mitochondrial/genetics , Histone Code , Humans , MicroRNAs/genetics , Recovery of Function , Treatment Outcome
20.
Oncogene ; 23(2): 524-34, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14724580

ABSTRACT

Rhabdomyosarcoma (RMS) tumors are the most common soft-tissue sarcomas in childhood. In this investigation, we show that myostatin, a skeletal muscle-specific inhibitor of growth and differentiation is expressed and translated in the cultured RMS cell line, RD. The addition of exogenous recombinant myostatin inhibits the proliferation of RD cells cultured in growth media, consistent with the role of myostatin in normal myoblast proliferation inhibition. However, unlike normal myoblasts, upregulation of p21 was not observed. Rather, myostatin signalling resulted in the specific downregulation of both Cdk2 and its cognate partner, cyclin-E. The analysis of Rb reveals that there was no change in its phosphorylation status with myostatin treatment, consistent with D-type-cyclin-Cdk4/6 complexes being active in the absence of p21. Moreover, the activity of Rb appeared to be unchanged between treated and nontreated RD cells, as determined by the ability of Rb to bind E2F1. The examination of NPAT, a substrate of cyclin-E-Cdk2 involved in the transcriptional activation of replication-dependent histone gene expression, revealed that it undergoes a loss of phosphorylation with myostatin treatment. Supporting this, a downregulation in H4-histone gene expression was observed. These results suggest that myostatin could potentially be used as an inhibitor of RMS proliferation and define a previously uncharacterized, Rb-independent mechanism for the inhibition of muscle precursor cell proliferation by myostatin.


Subject(s)
Proto-Oncogene Proteins , Retinoblastoma Protein/metabolism , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Transforming Growth Factor beta/pharmacology , CDC2-CDC28 Kinases/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cell Line, Tumor , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Histones/genetics , Humans , Myostatin , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Rhabdomyosarcoma/genetics
SELECTION OF CITATIONS
SEARCH DETAIL