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










Publication year range
1.
J Neuroinflammation ; 19(1): 246, 2022 Oct 05.
Article in English | MEDLINE | ID: mdl-36199097

ABSTRACT

Differential microglial inflammatory responses play a role in regulation of differentiation and maturation of oligodendrocytes (OLs) in brain white matter. How microglia-OL crosstalk is altered by traumatic brain injury (TBI) and its impact on axonal myelination and neurological function impairment remain poorly understood. In this study, we investigated roles of a Na+/H+ exchanger (NHE1), an essential microglial pH regulatory protein, in microglial proinflammatory activation and OL survival and differentiation in a murine TBI model induced by controlled cortical impact. Similar TBI-induced contusion volumes were detected in the Cx3cr1-CreERT2 control (Ctrl) mice and selective microglial Nhe1 knockout (Cx3cr1-CreERT2;Nhe1flox/flox, Nhe1 cKO) mice. Compared to the Ctrl mice, the Nhe1 cKO mice displayed increased resistance to initial TBI-induced white matter damage and accelerated chronic phase of OL regeneration at 30 days post-TBI. The cKO brains presented increased anti-inflammatory phenotypes of microglia and infiltrated myeloid cells, with reduced proinflammatory transcriptome profiles. Moreover, the cKO mice exhibited accelerated post-TBI sensorimotor and cognitive functional recovery than the Ctrl mice. These phenotypic outcomes in cKO mice were recapitulated in C57BL6J wild-type TBI mice receiving treatment of a potent NHE1 inhibitor HOE642 for 1-7 days post-TBI. Taken together, these findings collectively demonstrated that blocking NHE1 protein stimulates restorative microglial activation in oligodendrogenesis and neuroprotection, which contributes to accelerated brain repair and neurological function recovery after TBI.


Subject(s)
Brain Injuries, Traumatic , White Matter , Animals , Brain Injuries, Traumatic/metabolism , Disease Models, Animal , Mice , Mice, Inbred C57BL , Microglia/metabolism , Oligodendroglia , Recovery of Function
3.
Proc Natl Acad Sci U S A ; 117(8): 4418-4427, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32051249

ABSTRACT

In Alzheimer's disease (AD), human Tau is phosphorylated at S199 (hTau-S199-P) by the protein kinase glycogen synthase kinase 3ß (GSK3ß). HTau-S199-P mislocalizes to dendritic spines, which induces synaptic dysfunction at the early stage of AD. The AKT kinase, once phosphorylated, inhibits GSK3ß by phosphorylating it at S9. In AD patients, the abundance of phosphorylated AKT with active GSK3ß implies that phosphorylated AKT was unable to inactivate GSK3ß. However, the underlying mechanism of the inability of phosphorylated AKT to phosphorylate GSK3ß remains unknown. Here, we show that total AKT and phosphorylated AKT was sulfhydrated at C77 due to the induction of intracellular hydrogen sulfide (H2S). The increase in intracellular H2S levels resulted from the induction of the proinflammatory cytokine, IL-1ß, which is a pathological hallmark of AD. Sulfhydrated AKT does not interact with GSK3ß, and therefore does not phosphorylate GSK3ß. Thus, active GSK3ß phosphorylates Tau aberrantly. In a transgenic knockin mouse (AKT-KI+/+) that lacked sulfhydrated AKT, the interaction between AKT or phospho-AKT with GSK3ß was restored, and GSK3ß became phosphorylated. In AKT-KI+/+ mice, expressing the pathogenic human Tau mutant (hTau-P301L), the hTau S199 phosphorylation was ameliorated as GSK3ß phosphorylation was regained. This event leads to a decrease in dendritic spine loss by reducing dendritic localization of hTau-S199-P, which improves cognitive dysfunctions. Sulfhydration of AKT was detected in the postmortem brains from AD patients; thus, it represents a posttranslational modification of AKT, which primarily contributes to synaptic dysfunction in AD.


Subject(s)
Alzheimer Disease/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hydrogen Sulfide/metabolism , Proto-Oncogene Proteins c-akt/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Amino Acid Motifs , Animals , Brain/metabolism , Female , Glycogen Synthase Kinase 3 beta/genetics , Humans , Interleukin-1beta/metabolism , Male , Mice , Mice, Transgenic , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , tau Proteins/genetics
4.
J Neurosci ; 40(2): 424-446, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31694961

ABSTRACT

Persistent endoplasmic reticulum (ER) stress in neurons is associated with activation of inflammatory cells and subsequent neuroinflammation following traumatic brain injury (TBI); however, the underlying mechanism remains elusive. We found that induction of neuronal-ER stress, which was mostly characterized by an increase in phosphorylation of a protein kinase R-like ER kinase (PERK) leads to release of excess interferon (IFN)ß due to atypical activation of the neuronal-STING signaling pathway. IFNß enforced activation and polarization of the primary microglial cells to inflammatory M1 phenotype with the secretion of a proinflammatory chemokine CXCL10 due to activation of STAT1 signaling. The secreted CXCL10, in turn, stimulated the T-cell infiltration by serving as the ligand and chemoattractant for CXCR3+ T-helper 1 (Th1) cells. The activation of microglial cells and infiltration of Th1 cells resulted in white matter injury, characterized by impaired myelin basic protein and neurofilament NF200, the reduced thickness of corpus callosum and external capsule, and decline of mature oligodendrocytes and oligodendrocyte precursor cells. Intranasal delivery of CXCL10 siRNA blocked Th1 infiltration but did not fully rescue microglial activation and white matter injury after TBI. However, impeding PERK-phosphorylation through the administration of GSK2656157 abrogated neuronal induction of IFNß, switched microglial polarization to M2 phenotype, prevented Th1 infiltration, and increased Th2 and Treg levels. These events ultimately attenuated the white matter injury and improved anxiety and depressive-like behavior following TBI.SIGNIFICANCE STATEMENT A recent clinical study showed that human brain trauma patients had enhanced expression of type-1 IFN; suggests that type-1 IFN signaling may potentially influence clinical outcome in TBI patients. However, it was not understood how TBI leads to an increase in IFNß and whether induction of IFNß has any influence on neuroinflammation, which is the primary reason for morbidity and mortality in TBI. Our study suggests that induction of PERK phosphorylation, a characteristic feature of ER stress is responsible for an increase in neuronal IFNß, which, in turn, activates microglial cells and subsequently manifests the infiltration of T cells to induce neuroinflammation and subsequently white matter injury. Blocking PERK phosphorylation using GSK2656157 (or PERK knockdown) the whole cascade of neuroinflammation was attenuated and improved cognitive function after TBI.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Endoplasmic Reticulum Stress/physiology , Microglia/metabolism , T-Lymphocytes , White Matter/physiopathology , eIF-2 Kinase/metabolism , Animals , Female , Interferon-beta/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Signal Transduction/physiology , White Matter/injuries
5.
J Neurotrauma ; 36(23): 3284-3296, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31169064

ABSTRACT

An enduring deficit in neurogenesis largely contributes to the development of severe post-traumatic psychiatric disorders such as anxiety, depression, and memory impairment following traumatic brain injury (TBI); however, the mechanism remains obscure. Here we have shown that an imbalance in the generation of γ-aminobutyric acid (GABA)ergic and glutamatergic neurons due to aberrant induction of vesicular glutamate transporter 1 (vGlut1)-positive glutamatergic cells is responsible for impaired neuronal differentiation in the hippocampus following TBI. To elucidate the molecular mechanism, we found that TBI activates a transcription factor, Pax3, by increasing its acetylation status, and subsequently induces Ngn2 transcription. This event, in turn, augments the vGlut1-expressing glutamatergic neurons and accumulation of excess glutamate in the hippocampus that can affect neuronal differentiation. In our study the acetylation of Pax3 was increased due to loss of its interaction with a deacetylase, histone deacetylase 4 (HDAC4), which was downregulated after TBI. TBI-induced activation of GSK3ß was responsible for the degradation of HDAC4. We also showed that overexpression of HDAC4 before TBI reduces Pax3 acetylation by restoring an interaction between HDAC4 and Pax3 in the hippocampus. This event prevents the aberrant induction of vGlut1-positive glutamatergic neurons by decreasing the Ngn2 level and subsequently reinforces the balance between GABAergic and glutamatergic neurons following TBI. Further, we found that overexpression of HDAC4 in the hippocampus improves anxiety, depressive-like behavior, and memory functions following TBI.


Subject(s)
Brain Injuries, Traumatic/enzymology , Down-Regulation/physiology , Histone Deacetylases/metabolism , Mental Disorders/enzymology , Neurogenesis/physiology , Animals , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/psychology , Female , Male , Maze Learning/physiology , Mental Disorders/pathology , Mental Disorders/psychology , Mice , Mice, Inbred C57BL
7.
Free Radic Biol Med ; 134: 630-643, 2019 04.
Article in English | MEDLINE | ID: mdl-30790655

ABSTRACT

Traumatic Brain Injury (TBI) affects more than 1.7 million Americans each year and about 30% of TBI-patients having visual impairments. The loss of retinal ganglion cells (RGC) in the retina and axonal degeneration in the optic nerve have been attributed to vision impairment following TBI; however, the molecular mechanism has not been elucidated. Here we have shown that an increase in histone di-methylation at lysine 9 residue (H3K9Me2), synthesized by the catalytic activity of a histone methyltransferase, G9a is responsible for RGC loss and axonal degeneration in the optic nerve following TBI. To elucidate the molecular mechanism, we found that an increase in H3K9Me2 results in the induction of oxidative stress both in the RGC and optic nerve by decreasing the mRNA level of antioxidants such as Superoxide dismutase (sod) and catalase through impairing the transcriptional activity of Nuclear factor E2-related factor 2 (Nrf2) via direct interaction. The induction of oxidative stress is associated with death in RGC and oligodendrocyte precursor cells (OPCs). The death in OPCs is correlated with a reduction in myelination, and the expression of myelin binding protein (MBP) in association with degeneration of neurofilaments in the optic nerve. This event allied to an impairment of the retrograde transport of axons and loss of nerve fiber layer in the optic nerve following TBI. An administration of G9a inhibitor, UNC0638 attenuates the induction of H3K9Me2 both in RGC and optic nerve and subsequently activates Nrf2 to reduce oxidative stress. This event was concomitant with the rescue in the loss of retinal thickness, attenuation in optic nerve degeneration and improvement in the retrograde transport of axons following TBI.


Subject(s)
Brain Injuries, Traumatic/complications , Histones/metabolism , Lysine/metabolism , Optic Nerve/pathology , Oxidative Stress , Retinal Ganglion Cells/pathology , Vision Disorders/etiology , Animals , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Histones/genetics , Lysine/genetics , Male , Methylation , Mice , Mice, Inbred C57BL , Myelin Sheath , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Optic Nerve/metabolism , Retinal Ganglion Cells/metabolism , Vision Disorders/metabolism , Vision Disorders/pathology
8.
Mech Ageing Dev ; 178: 72-79, 2019 03.
Article in English | MEDLINE | ID: mdl-30668956

ABSTRACT

Tau, a microtubule-associated protein promotes assembly and stability of microtubules which is related to axoplasmic flow and critical neuronal activities upon physiological conditions. Under neurodegenerative condition such as in Alzheimer's Disease (AD), tau-microtubule binding dynamics and equilibrium are severely affected due to its aberrant post-translational modifications including acetylation and hyperphosphorylation. This event results in its conformational changes to form neurofibrillary tangles (NFT) after aggregation in the cytosol. The formation of NFT is more strongly correlated with cognitive decline than the distribution of senile plaque, which is formed by polymorphous beta-amyloid (Aß) protein deposits, another pathological hallmark of AD. In neurodegenerative conditions, other than AD, the disease manifestation is correlated with mutations of the MAPT gene. In Primary age-related tauopathy (PART), which is commonly observed in the brains of aged individuals, tau deposition is directly correlated with cognitive deficits even in the absence of Aß deposition. Thus, tauopathy has been considered as an essential hallmark in neurodegeneration and normal brain aging. In this review, we highlighted the recent progress about the tauopathies in the light of its posttranslational modifications and its implication in AD and the aged brain.


Subject(s)
Aging/metabolism , Brain/metabolism , Protein Processing, Post-Translational , Tauopathies/metabolism , tau Proteins/metabolism , Acetylation , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/pathology , Cognitive Dysfunction/metabolism , Humans , Neurofibrillary Tangles/metabolism , Oxidative Stress/physiology , Phosphorylation , Plaque, Amyloid/physiopathology , Tauopathies/pathology
9.
Neuroscientist ; 25(5): 420-433, 2019 10.
Article in English | MEDLINE | ID: mdl-30477403

ABSTRACT

The brain undergoes several changes at structural, molecular, and cellular levels leading to alteration in its functions and these processes are primarily maintained by proteostasis in cells. However, an imbalance in proteostasis due to the abnormal accumulation of protein aggregates induces endoplasmic reticulum (ER) stress. This event, in turn, activate the unfolded protein response; however, in most neurodegenerative conditions and brain injury, an uncontrolled unfolded protein response elicits memory dysfunction. Although the underlying signaling mechanism for impairment of memory function following induction of ER stress remains elusive, recent studies have highlighted that inactivation of a transcription factor, CREB, which is essential for synaptic function and memory formation, plays an essential role for ER stress-induced synaptic and memory dysfunction. In this review, current studies and most updated view on how ER stress affects memory function in both physiological and pathological conditions will be highlighted.


Subject(s)
Brain Injuries/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Endoplasmic Reticulum Stress/physiology , Memory/physiology , Proteostasis/physiology , Unfolded Protein Response/physiology , Animals , Brain Injuries/complications , Humans , Memory Disorders/etiology , Memory Disorders/metabolism , Neurons/metabolism , Signal Transduction
10.
Neurobiol Dis ; 118: 108-116, 2018 10.
Article in English | MEDLINE | ID: mdl-30010002

ABSTRACT

Cell cycle activation has been associated with varying types of neurological disorders including brain injury. Cyclin D1 is a critical modulator of cell cycle activation and upregulation of Cyclin D1 in neurons contributes to the pathology associated with traumatic brain injury (TBI). Mitochondrial mass is a critical factor to maintain the mitochondrial function, and it can be regulated by different signaling cascades and transcription factors including NRF1. However, the underlying mechanism of how TBI leads to impairment of mitochondrial mass following TBI remains obscure. Our results indicate that augmentation of CyclinD1 attenuates mitochondrial mass formation following TBI. To elucidate the molecular mechanism, we found that Cyclin D1 interacts with a transcription factor NRF1 in the nucleus and prevents NRF1's interaction with p300 in the pericontusional cortex following TBI. As a result, the acetylation level of NRF1 was decreased, and its transcriptional activity was attenuated. This event leads to a loss of mitochondrial mass in the pericontusional cortex following TBI. Intranasal delivery of Cyclin D1 RNAi immediately after TBI rescues transcriptional activation of NRF1 and recovers mitochondrial mass after TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cyclin D1/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Animals , Brain Injuries, Traumatic/genetics , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/genetics , RNA Interference/physiology
11.
Sci Signal ; 11(522)2018 03 20.
Article in English | MEDLINE | ID: mdl-29559585

ABSTRACT

Acetylation of the microtubule-associated protein tau promotes its polymerization into neurofibrillary tangles that are implicated in the pathology of Alzheimer's disease (AD). The gaseous neurotransmitter nitric oxide (NO) regulates cell signaling through the nitrosylation of proteins. We found that NO production and tau acetylation at Lys280 occurred in the brain tissue in mice and in cultured mouse cortical neurons in response to exposure to amyloid-ß1-42 (Aß1-42), a peptide that is also implicated in AD. An increased abundance of NO facilitated the S-nitrosylation (SNO) of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). S-nitrosylated GAPDH (GAPDH-SNO) promoted the acetylation and activation of the acetyltransferase p300 and facilitated the nitrosylation and inactivation of the deacetylase sirtuin 1 (SIRT1). The abundance of GAPDH-SNO was increased in postmortem brain samples from AD patients. Preventing the increase in GAPDH-SNO abundance in both cultured neurons and mice, either by overexpression of the nitrosylation mutant of GAPDH (GAPDH C150S) or by treatment with the GAPDH nitrosylation inhibitor CGP3466B (also known as omigapil), abrogated Aß1-42-induced tau acetylation, memory impairment, and locomotor dysfunction in mice, suggesting that this drug might be repurposed to treat patients with AD.


Subject(s)
Alzheimer Disease/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Nitric Oxide Synthase Type I/deficiency , Nitric Oxide/metabolism , Sirtuin 1/metabolism , p300-CBP Transcription Factors/metabolism , tau Proteins/metabolism , Acetylation/drug effects , Alzheimer Disease/chemically induced , Alzheimer Disease/genetics , Amyloid beta-Peptides/toxicity , Animals , Brain/drug effects , Brain/metabolism , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Nitric Oxide Synthase Type I/genetics , Oxepins/pharmacology , Peptide Fragments/toxicity
12.
J Neurosci ; 37(24): 5900-5911, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28522733

ABSTRACT

The PKR-like ER kinase (PERK), a transmembrane protein, resides in the endoplasmic reticulum (ER). Its activation serves as a key sensor of ER stress, which has been implicated in traumatic brain injury (TBI). The loss of memory is one of the most common symptoms after TBI, but the precise role of PERK activation in memory impairment after TBI has not been well elucidated. Here, we have shown that blocking the activation of PERK using GSK2656157 prevents the loss of dendritic spines and rescues memory deficits after TBI. To elucidate the molecular mechanism, we found that activated PERK phosphorylates CAMP response element binding protein (CREB) and PSD95 directly at the S129 and T19 residues, respectively. Phosphorylation of CREB protein prevents its interaction with a coactivator, CREB-binding protein, and subsequently reduces the BDNF level after TBI. Conversely, phosphorylation of PSD95 leads to its downregulation in pericontusional cortex after TBI in male mice. Treatment with either GSK2656157 or overexpression of a kinase-dead mutant of PERK (PERK-K618A) rescues BDNF and PSD95 levels in the pericontusional cortex by reducing phosphorylation of CREB and PSD95 proteins after TBI. Similarly, administration of either GSK2656157 or overexpression of PERK-K618A in primary neurons rescues the loss of dendritic outgrowth and number of synapses after treatment with a PERK activator, tunicamycin. Therefore, our study suggests that inhibition of PERK phosphorylation could be a potential therapeutic target to restore memory deficits after TBI.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is the leading cause of death and disability around the world and affects 1.7 million Americans each year. Here, we have shown that TBI-activated PKR-like ER kinase (PERK) is responsible for memory deficiency, which is the most common problem in TBI patients. A majority of PERK's biological activities have been attributed to its function as an eIF2α kinase. However, our study suggests that activated PERK mediates its function via increasing phosphorylation of CAMP response element binding protein (CREB) and PSD95 after TBI. Blocking PERK phosphorylation rescues spine loss and memory deficits independently of phosphorylation of eIF2α. Therefore, our study suggests that CREB and PSD95 are novel substrates of PERK, so inhibition of PERK phosphorylation using GSK2656157 would be beneficial against memory impairment after TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Guanylate Kinases/metabolism , Membrane Proteins/metabolism , Memory Disorders/metabolism , eIF-2 Kinase/metabolism , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Dendritic Spines/pathology , Disks Large Homolog 4 Protein , Down-Regulation , Enzyme Activation , Male , Memory Disorders/etiology , Memory Disorders/pathology , Mice , Mice, Inbred C57BL
13.
Neurochem Int ; 111: 103-107, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28163060

ABSTRACT

Traumatic brain injury (TBI) is one of the major cause of morbidity and mortality and it affects more than 1.7 million Americans each year. Depending on its location and severity, TBI leads to structural and functional damage in several parts of the brain such as cranial nerves, optic nerve tract or other circuitry involved in vision, and occipital lobe. As a result, the function associated with vision processing and perception are significantly affected and cause blurred vision, double vision, decreased peripheral vision and blindness. In this mini-review, we will focus the recent progress made to understand the pathology and underlying cellular/molecular mechanisms involved in the impairment of the integrity of visual systems following TBI.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Brain/physiopathology , Optic Nerve Injuries/physiopathology , Optic Nerve/physiopathology , Animals , Cognition/physiology , Humans , Vision, Ocular/physiology
14.
J Mol Biol ; 429(4): 543-561, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28013031

ABSTRACT

Hydrogen sulfide (H2S), a novel gasotransmitter, is endogenously synthesized by multiple enzymes that are differentially expressed in the peripheral tissues and central nervous systems. H2S regulates a wide range of physiological processes, namely cardiovascular, neuronal, immune, respiratory, gastrointestinal, liver, and endocrine systems, by influencing cellular signaling pathways and sulfhydration of target proteins. This review focuses on the recent progress made in H2S signaling that affects mechanistic and functional aspects of several biological processes such as autophagy, inflammation, proliferation and differentiation of stem cell, cell survival/death, and cellular metabolism under both physiological and pathological conditions. Moreover, we highlighted the cross-talk between nitric oxide and H2S in several bilogical contexts.


Subject(s)
Gasotransmitters/metabolism , Hydrogen Sulfide/metabolism , Neurotransmitter Agents/metabolism , Signal Transduction , Animals , Brain Injuries, Traumatic/pathology , Cell Cycle , Cell Differentiation , Cell Proliferation , Fibrosis/pathology , Humans , Ischemia/pathology , Metabolic Diseases/pathology , Neurodegenerative Diseases/metabolism , Nitric Oxide/metabolism , Stem Cells/cytology
15.
Oncotarget ; 7(48): 78220-78221, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27829240
16.
Neurobiol Dis ; 96: 12-21, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27544482

ABSTRACT

Anesthetics including isoflurane are known to induce neuronal dysfunction in the developing brain, however, the underlying mechanism is mostly unknown. The transcriptional activation of CREB (cyclic AMP response element binding protein) and the alterations in acetylation of histones modulated by several histone deacetylases such as HDAC4 (histone deacetylase 4) are known to contribute to synaptic plasticity in the brain. Here we have shown that administration of isoflurane (1.4%) for 2h leads to transcriptional inactivation of CREB which results in loss of dendritic outgrowth and decreased expression level of proteins essential for memory and cognitive functions, such as BDNF, and c-fos in the developing brain of mice at postnatal day 7 (PND7). To elucidate the molecular mechanism, we found that exposure to isoflurane leads to an increase in nuclear translocation of HDAC4, which interacts with CREB in the nucleus. This event, in turn, results in a decrease in interaction between an acetyltransferase, CBP, and CREB that ultimately leads to transcriptional inactivation of CREB. As a result, the expression level of BDNF, and c-fos were significantly down-regulated after administration of isoflurane in PND7 brain. Depletion of HDAC4 in PND7 brain rescues the transcriptional activation of CREB along with augmentation in the level of the expression level of BDNF and c-fos. Moreover, administration of lentiviral particles of HDAC4 RNAi in primary neurons rescues neurite outgrowth following isoflurane treatment. Taken together, our study suggests that HDAC4-induced transcriptional inactivation of CREB is responsible for isoflurane-induced cognitive dysfunction in the brain.


Subject(s)
Anesthetics, Inhalation/toxicity , Brain , CREB-Binding Protein/metabolism , Cognition Disorders/chemically induced , Histone Deacetylases/metabolism , Isoflurane/toxicity , Animals , Animals, Newborn , Brain/drug effects , Brain/growth & development , Brain/metabolism , Brain/pathology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Disease Models, Animal , Down-Regulation/drug effects , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Histone Deacetylases/genetics , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neuronal Outgrowth/drug effects , Neurons/cytology , Neurons/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Reflex, Righting/drug effects
17.
Neuropharmacology ; 107: 79-88, 2016 08.
Article in English | MEDLINE | ID: mdl-26970014

ABSTRACT

Traumatic brain injury (TBI) is one of the major cause of morbidity and mortality and it affects more than 1.7 million people in the USA. A couple of regenerative pathways including activation of hypoxia-inducible transcription factor 1 alpha (HIF-1α) are initiated to reduce cellular damage following TBI; however endogenous activation of these pathways is not enough to provide neuroprotection after TBI. Thus we aimed to see whether sustained activation of HIF-1α can provide neuroprotection and neurorepair following TBI. We found that chronic treatment with dimethyloxaloylglycine (DMOG) markedly increases the expression level of HIF-1α and mRNA levels of its downstream proteins such as Vascular endothelial growth factor (VEGF), Phosphoinositide-dependent kinase-1 and 4 (PDK1, PDK4) and Erythropoietin (EPO). Treatment of DMOG activates a major cell survival protein kinase Akt and reduces both cell death and lesion volume following TBI. Moreover, administration of DMOG augments cluster of differentiation 31 (CD31) staining in pericontusional cortex after TBI, which suggests that DMOG stimulates angiogenesis after TBI. Treatment with DMOG also improves both memory and motor functions after TBI. Taken together our results suggest that sustained activation of HIF-1α provides significant neuroprotection following TBI.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Brain Injuries, Traumatic/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neuroprotective Agents/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Angiogenesis Inducing Agents/pharmacology , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/psychology , Cell Death/drug effects , Cell Death/physiology , Disease Models, Animal , Erythropoietin/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/metabolism , Memory Disorders/pathology , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Nootropic Agents/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism
18.
Rev Neurosci ; 27(1): 93-100, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26352199

ABSTRACT

Traumatic brain injury (TBI), a major global health and socioeconomic problem, is now established as a chronic disease process with a broad spectrum of pathophysiological symptoms followed by long-term disabilities. It triggers multiple and multidirectional biochemical events that lead to neurodegeneration and cognitive impairment. Recent studies have presented strong evidence that patients with TBI history have a tendency to develop proteinopathy, which is the pathophysiological feature of neurodegenerative disorders such as Alzheimer disease (AD), chronic traumatic encephalopathy (CTE), and amyotrophic lateral sclerosis (ALS). This review mainly focuses on mechanisms related to AD, CTE, and ALS that are induced after TBI and their relevance to the advancement of these neurodegenerative diseases. This review encompasses acute effects and chronic neurodegenerative consequences after TBI for a better understanding of TBI-induced neuronal death and to design therapies that will effectively treat patients in the primary or secondary progressive stages.


Subject(s)
Alzheimer Disease/etiology , Amyotrophic Lateral Sclerosis/etiology , Brain Injuries/complications , Alzheimer Disease/epidemiology , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/epidemiology , Amyotrophic Lateral Sclerosis/metabolism , Animals , Brain Injuries/metabolism , Humans
19.
Neuromolecular Med ; 17(2): 97-110, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24777294

ABSTRACT

Synaptic plasticity is one of the most fundamental properties of neurons that underlie the formation of the memory in brain. In recent years, epigenetic modification of both DNA and histones such as DNA methylation and histone acetylation and methylation emerges as a potential regulatory mechanism that governs the transcription of several genes responsible for memory formation and behavior. Furthermore, the recent identification of nitrosylation of proteins has shown to either activate or repress gene transcription by modulating histone methylation or acetylation status in mature neuron. Recent studies suggest that the use of major substrates of abuse, e.g., cocaine, induces alterations in molecular and cellular mechanisms of epigenetics that underlie long-term memories in the striatum and prefrontal cortex. Moreover, downregulation of genes due to alterations in epigenetics leads to cognitive deficiencies associated with neurological disorders such as Alzheimer's disease, Huntington's disease, psychiatric disorder such as Rett's syndrome and aging. In this review, I will discuss the evidence for several epigenetic mechanisms in the coordination of complex memory formation and storage. In addition, I will address the current literature highlighting the role of acetylation and methylation of chromatin in memory impairment associated with several neurological disorders, aging, and addiction.


Subject(s)
Chromatin/genetics , Epigenesis, Genetic , Memory/physiology , Acetylation , Aging/genetics , Aging/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , DNA Methylation , Histone Code , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/physiology , Histones/metabolism , Humans , Methylation , Nerve Growth Factors/physiology , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Neuronal Plasticity/genetics , Nitric Oxide/physiology , Nitrosation , Protein Processing, Post-Translational , Substance-Related Disorders/genetics , Substance-Related Disorders/metabolism
20.
Mol Cell ; 56(6): 786-95, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25435139

ABSTRACT

Induction of a proinflammatory cytokine, interleukin-1ß (IL-1ß) plays a role in memory impairment associated with various neurological disorders and brain injury. Here we show that IL-1ß-induced memory impairment in brain is mediated by hydrogen sulfide (H2S) synthesized by cystathionine beta-synthase (CBS). H2S modifies GAPDH essentially via sulfhydration in dendrites, which promotes its binding to the E3 ligase protein, Siah. Then Siah binds to a critical synaptic scaffolding molecule, PSD95, and leads it to degradation via ubiquitination. In CBS heterozygous mice (cbs(+/-)) and primary neurons depleted with either CBS or IL-1R, IL-1ß-induced loss of PSD95 was rescued along with a decrease in the level of GAPDH sulfhydration. Moreover, decrease in the loss of PSD95 in cbs(+/-) mice results in improvement of IL-1ß-induced cognitive deficits and neurobehavioral outcomes. Thus, our findings reveal a mechanism where GAPDH sulfhydration appears to be a physiologic determinant of cytokine-induced memory impairment in brain.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Guanylate Kinases/metabolism , Interleukin-1beta/physiology , Membrane Proteins/metabolism , Animals , Cystathionine beta-Synthase/metabolism , Disks Large Homolog 4 Protein , HEK293 Cells , Humans , Hydrogen Sulfide/metabolism , Male , Memory Disorders/enzymology , Mice, Inbred C57BL , Mice, Transgenic , Protein Processing, Post-Translational , Proteolysis , Synapses/enzymology , Synapses/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...