Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Int J Mol Sci ; 24(6)2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36982673

ABSTRACT

We launched our Special Issue (SI) at the beginning of 2021, with the hope to bring together current research in the field of neurodegeneration [...].


Subject(s)
Nervous System Diseases , Humans , Nervous System Diseases/therapy
3.
Brain Sci ; 12(2)2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35203953

ABSTRACT

Ever since it was first reported in Wuhan, China, the coronavirus-induced disease of 2019 (COVID-19) has become an enigma of sorts with ever expanding reports of direct and indirect effects of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on almost all the vital organ systems. Along with inciting acute pulmonary complications, the virus attacks the cardiac, renal, hepatic, and gastrointestinal systems as well as the central nervous system (CNS). The person-to-person variability in susceptibility of individuals to disease severity still remains a puzzle, although the comorbidities and the age/gender of a person are believed to play a key role. SARS-CoV-2 needs angiotensin-converting enzyme 2 (ACE2) receptor for its infectivity, and the association between SARS-CoV-2 and ACE2 leads to a decline in ACE2 activity and its neuroprotective effects. Acute respiratory distress may also induce hypoxia, leading to increased oxidative stress and neurodegeneration. Infection of the neurons along with peripheral leukocytes' activation results in proinflammatory cytokine release, rendering the brain more susceptible to neurodegenerative changes. Due to the advancement in molecular biology techniques and vaccine development programs, the world now has hope to relatively quickly study and combat the deadly virus. On the other side, however, the virus seems to be still evolving with new variants being discovered periodically. In keeping up with the pace of this virus, there has been an avalanche of studies. This review provides an update on the recent progress in adjudicating the CNS-related mechanisms of SARS-CoV-2 infection and its potential to incite or accelerate neurodegeneration in surviving patients. Current as well as emerging therapeutic opportunities and biomarker development are highlighted.

4.
Int J Mol Sci ; 22(21)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34768950

ABSTRACT

Alzheimer's disease (AD) is a major cause of dementia in older adults and is fast becoming a major societal and economic burden due to an increase in life expectancy. Age seems to be the major factor driving AD, and currently, only symptomatic treatments are available. AD has a complex etiology, although mitochondrial dysfunction, oxidative stress, inflammation, and metabolic abnormalities have been widely and deeply investigated as plausible mechanisms for its neuropathology. Aß plaques and hyperphosphorylated tau aggregates, along with cognitive deficits and behavioral problems, are the hallmarks of the disease. Restoration of mitochondrial bioenergetics, prevention of oxidative stress, and diet and exercise seem to be effective in reducing Aß and in ameliorating learning and memory problems. Many mitochondria-targeted antioxidants have been tested in AD and are currently in development. However, larger streamlined clinical studies are needed to provide hard evidence of benefits in AD. This review discusses the causative factors, as well as potential therapeutics employed in the treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Mitochondria/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Animals , Antioxidants/pharmacology , Diet , Disease Models, Animal , Energy Metabolism , Exercise , Glucose/metabolism , Humans , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Models, Neurological , Neurons/drug effects , Neurons/metabolism , Oxidative Stress , tau Proteins/metabolism
5.
Brain Sci ; 11(11)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34827436

ABSTRACT

Mitochondria associated membranes (MAMs), as the name suggests, are the membranes that physically and biochemically connect mitochondria with endoplasmic reticulum. MAMs not only structurally but also functionally connect these two important organelles within the cell which were previously thought to exist independently. There are multiple points of communication between ER-mitochondria and MAMs play an important role in both ER and mitochondria functions such as Ca2+ homeostasis, proteostasis, mitochondrial bioenergetics, movement, and mitophagy. The number of disease-related proteins and genes being associated with MAMs has been continually on the rise since its discovery. There is an overwhelming overlap between the biochemical functions of MAMs and processes affected in neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Thus, MAMs have received well-deserving and much delayed attention as modulators for ER-mitochondria communication and function. This review briefly discusses the recent progress made in this now fast developing field full of promise for very exciting future therapeutic discoveries.

6.
Hum Mol Genet ; 25(11): 2269-2282, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27008868

ABSTRACT

Huntington's disease (HD) is a devastating illness and at present there is no disease modifying therapy or cure for it; and management of the disease is limited to a few treatment options for amelioration of symptoms. Recently, we showed that the administration of bezafibrate, a pan-PPAR agonist, increases the expression of PGC-1α and mitochondrial biogenesis, and improves phenotype and survival in R6/2 transgenic mouse model of HD. Since the R6/2 mice represent a 'truncated' huntingtin (Htt) mouse model of HD, we tested the efficacy of bezafibrate in a 'full-length' Htt mouse model, the BACHD mice. Bezafibrate treatment restored the impaired PPARγ, PPARδ, PGC-1α signaling pathway, enhanced mitochondrial biogenesis and improved antioxidant defense in the striatum of BACHD mice. Untreated BACHD mice show robust and progressive motor deficits, as well as late-onset and selective neuropathology in the striatum, which was markedly ameliorated in the BACHD mice treated with bezafibrate. Our data demonstrate the efficacy of bezafibrate in ameliorating both neuropathological features and disease phenotype in BACHD mice, and taken together with our previous studies with the R6/2 mice, highlight the strong therapeutic potential of bezafibrate for treatment of HD.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/drug therapy , PPAR delta/biosynthesis , PPAR gamma/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Animals , Bezafibrate/administration & dosage , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/genetics , Organelle Biogenesis , PPAR delta/genetics , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/antagonists & inhibitors , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Signal Transduction/drug effects
7.
Mov Disord ; 29(3): 285-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24573776

ABSTRACT

Huntington's disease (HD) is a prototypical dominantly inherited neurodegenerative disorder characterized by progressive cognitive deterioration, psychiatric disturbances, and a movement disorder. The genetic cause of the illness is a CAG repeat expansion in the huntingtin gene, which leads to a polyglutamine expansion in the huntingtin protein. The exact mechanism by which mutant huntingtin causes HD is unknown, but it causes abnormalities in gene transcription as well as both mitochondrial dysfunction and oxidative damage. Because the penetrance of HD is complete with CAG repeats greater than 39, patients can be diagnosed well before disease onset with genetic testing. Longitudinal studies of HD patients before disease onset have shown that subtle cognitive and motor deficits occur as much as 10 years before onset, as do reductions in glucose utilization and striatal atrophy. An increase in inflammation, as shown by elevated interleukin-6, occurs approximately 15 years before onset. Detection of these abnormalities may be useful in defining an optimal time for disease intervention to try to slow or halt the degenerative process. Although reducing gene expression with small interfering RNA or short hairpin RNA is an attractive approach, other approaches targeting energy metabolism, inflammation, and oxidative damage may be more easily and rapidly moved into the clinic. The recent PREQUEL study of coenzyme Q10 in presymptomatic gene carriers showed the feasibility of carrying out clinical trials to slow or halt onset of HD. We review both the earliest detectable clinical and laboratory manifestations of HD, as well as potential neuroprotective therapies that could be utilized in presymptomatic HD.


Subject(s)
Huntington Disease/genetics , Huntington Disease/therapy , Mitochondria/genetics , Neuroprotective Agents/therapeutic use , Ubiquinone/analogs & derivatives , Animals , Disease Models, Animal , Genetic Testing , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Ubiquinone/drug effects , Ubiquinone/metabolism
8.
Free Radic Biol Med ; 62: 37-46, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23602910

ABSTRACT

The constant high energy demand of neurons makes them rely heavily on their mitochondria. Dysfunction of mitochondrial energy metabolism leads to reduced ATP production, impaired calcium buffering, and generation of reactive oxygen species. There is strong evidence that mitochondrial dysfunction results in neurodegeneration and may contribute to the pathogenesis of Huntington's disease (HD). Studies over the past few years have implicated an impaired function of peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1α (PGC-1α), a transcriptional master coregulator of mitochondrial biogenesis, metabolism, and antioxidant defenses, in causing mitochondrial dysfunction in HD. Here we have attempted to discuss in a nutshell, the key findings on the role of PGC-1α in mitochondrial dysfunction in HD and its potential as a therapeutic target to cure HD.


Subject(s)
Huntington Disease/metabolism , Mitochondria/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Adenosine Triphosphate/biosynthesis , Antioxidants/metabolism , Calcium/metabolism , Energy Metabolism , Humans , Huntington Disease/pathology , Mitochondria/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Trans-Activators/metabolism
9.
J Pharmacol Exp Ther ; 342(3): 619-30, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22700435

ABSTRACT

Neurodegenerative diseases are a large group of disabling disorders of the nervous system, characterized by the relative selective death of neuronal subtypes. In most cases, there is overwhelming evidence of impaired mitochondrial function as a causative factor in these diseases. More recently, evidence has emerged for impaired mitochondrial dynamics (shape, size, fission-fusion, distribution, movement etc.) in neurodegenerative diseases such as Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Alzheimer's disease. Here, we provide a concise overview of the major findings in recent years highlighting the importance of healthy mitochondria for a healthy neuron.


Subject(s)
Mitochondria/pathology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neurons/pathology , Neurons/physiology , Animals , Humans , Mitochondrial Dynamics/physiology
10.
PLoS One ; 7(5): e36191, 2012.
Article in English | MEDLINE | ID: mdl-22563483

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease in which the etiology of 90 percent of the patients is unknown. Pesticide exposure is a major risk factor for PD, and paraquat (PQ), pyridaben (PY) and maneb (MN) are amongst the most widely used pesticides. We studied mRNA expression using transcriptome sequencing (RNA-Seq) in the ventral midbrain (VMB) and striatum (STR) of PQ, PY and paraquat+maneb (MNPQ) treated mice, followed by pathway analysis. We found concordance of signaling pathways between the three pesticide models in both the VMB and STR as well as concordance in these two brain areas. The concordant signaling pathways with relevance to PD pathogenesis were e.g. axonal guidance signaling, Wnt/ß-catenin signaling, as well as pathways not previously linked to PD, e.g. basal cell carcinoma, human embryonic stem cell pluripotency and role of macrophages, fibroblasts and endothelial cells in rheumatoid arthritis. Human PD pathways previously identified by expression analysis, concordant with VMB pathways identified in our study were axonal guidance signaling, Wnt/ß-catenin signaling, IL-6 signaling, ephrin receptor signaling, TGF-ß signaling, PPAR signaling and G-protein coupled receptor signaling. Human PD pathways concordant with the STR pathways in our study were Wnt/ß-catenin signaling, axonal guidance signaling and G-protein coupled receptor signaling. Peroxisome proliferator activated receptor delta (Ppard) and G-Protein Coupled Receptors (GPCRs) were common genes in VMB and STR identified by network analysis. In conclusion, the pesticides PQ, PY and MNPQ elicit common signaling pathways in the VMB and STR in mice, which are concordant with known signaling pathways identified in human PD, suggesting that these pathways contribute to the pathogenesis of idiopathic PD. The analysis of these networks and pathways may therefore lead to improved understanding of disease pathogenesis, and potential novel therapeutic targets.


Subject(s)
Parkinson Disease/genetics , Pesticides/toxicity , Signal Transduction/genetics , Transcriptome/drug effects , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Gene Expression Profiling , Gene Regulatory Networks/drug effects , Humans , Male , Maneb/toxicity , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Paraquat/toxicity , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/genetics , Pyridazines/toxicity , Reverse Transcriptase Polymerase Chain Reaction
11.
Hum Mol Genet ; 21(15): 3474-88, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22589249

ABSTRACT

Huntington's disease (HD) is an incurable neurological disorder caused by an abnormal glutamine repeat expansion in the huntingtin (Htt) protein. In the present studies, we investigated the role of Transducers of Regulated cAMP response element-binding (CREB) protein activity (TORCs) in HD, since TORCs play an important role in the expression of the transcriptional co-regulator peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), whose expression is impaired in HD. We found significantly decreased TORC1 expression levels in STHdhQ111 cells expressing mutant Htt, in the striatum of NLS-N171-82Q, R6/2 and HdhQ111 HD transgenic mice and in postmortem striatal tissue from HD patients. TORC1 overexpression in wild-type (WT) and Htt striatal cells increased CREB mRNA and protein levels, PGC-1α promoter activity, mRNA expression of the PGC-1α, NRF-1, Tfam and CytC genes, mitochondrial DNA content, mitochondrial activity and mitochondrial membrane potential. TORC1 overexpression also increased the resistance of striatal cells to 3-nitropropionic (3-NP) acid-mediated toxicity. In cultured WT and mutant Htt striatal cells, small hairpin RNA-mediated TORC1 knockdown resulted in decreased PGC-1α expression and increased susceptibility to 3-NP-induced toxicity. Overexpression of PGC-1α partially prevented TORC1 knockdown-mediated increased susceptibility of Htt striatal cells to 3-NP. Specific knockdown of TORC1 in the striatum of NLS-N171-82Q HD transgenic mice induced neurodegeneration. Lastly, knockdown of Htt prevents transcriptional repression of TORC1 and CREB in Htt striatal cells. These findings show that impaired expression and function of TORC1, which results in a reduction in PGC-1α, plays an important role in mitochondrial dysfunction in HD.


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Huntingtin Protein , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic
12.
Cell Metab ; 15(5): 567-9, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22560208

ABSTRACT

Impaired activity of peroxisome proliferator-activated receptor (PPAR)-γ coactivator (PGC)-1α has been implicated in the pathophysiology of several neurodegenerative disorders. In this issue, Da Cruz et al. (2012) show improved muscle function, but not survival, with increased PGC-1α activity in muscle in a mouse model of amyotrophic lateral sclerosis.

13.
Biochim Biophys Acta ; 1822(5): 664-74, 2012 May.
Article in English | MEDLINE | ID: mdl-22138129

ABSTRACT

Huntington's disease (HD) is a prototypical neurodegenerative disease in which there is selective neuronal degeneration, which leads to progressive disability, manifesting itself as a movement disorder, with both psychiatric and cognitive impairment. The disease is caused by a cytosine-adenine-guanine (CAG) repeat expansion in the huntingtin gene, which causes an expanded polyglutamine repeat in the huntingtin protein, resulting in a protein with a novel gain of function. The mutant huntingtin protein causes neuronal dysfunction and eventual cell death in which transcriptional impairment, excitotoxicity, oxidative damage, inflammation, apoptosis and mitochondrial dysfunction are all implicated. A critical transcriptional impairment may be impaired expression and function of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), a master co-regulator of mitochondrial biogenesis and expression of antioxidant enzymes. A deficiency of PGC-1α leads to increased vulnerability to oxidative stress and to striatal degeneration. The extent and severity of the oxidative damage in HD are features well recognized but perhaps under-appreciated. Oxidative damage occurs to lipids, proteins and deoxyribonucleic acid (DNA), and it has been suggested that the latter may contribute to CAG repeat expansion during DNA repair [1]. A marked elevation of oxidized DNA bases occurs in patients' plasma, which may provide a biomarker of disease progression. Antioxidants are effective in slowing disease progression in transgenic mouse models of HD, and show promise in human clinical trials. Strategies to transcriptionally increase expression of antioxidant enzymes by modulating the Nrf-2/ARE pathway, or by increasing expression of PGC-1α hold great promise for developing new treatments to slow or halt the progression of HD. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.


Subject(s)
Antioxidants/therapeutic use , Huntington Disease/drug therapy , Humans , Huntington Disease/blood , Huntington Disease/genetics , Oxidative Stress , Reactive Oxygen Species/metabolism
14.
Hum Mol Genet ; 21(5): 1124-37, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22095692

ABSTRACT

There is substantial evidence that impairment of peroxisome proliferator-activated receptor (PPAR)-γ-coactivator 1α (PGC-1α) levels and activity play an important role in Huntington's disease (HD) pathogenesis. We tested whether pharmacologic treatment with the pan-PPAR agonist bezafibrate would correct a deficiency of PGC-1α and exert beneficial effects in a transgenic mouse model of HD. We found that administration of bezafibrate in the diet restored levels of PGC-1α, PPARs and downstream genes to levels which occur in wild-type mice. There were significant improvements in phenotype and survival. In the striatum, astrogliosis and neuronal atrophy were attenuated and numbers of mitochondria were increased. Bezafibrate treatment prevented conversion of type I oxidative to type II glycolytic muscle fibers and increased the numbers of muscle mitochondria. Finally, bezafibrate rescued lipid accumulation and apparent vacuolization of brown adipose tissue in the HD mice. These findings provide strong evidence that treatment with bezafibrate exerts neuroprotective effects which may be beneficial in the treatment of HD.


Subject(s)
Bezafibrate/pharmacology , Huntington Disease/drug therapy , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Trans-Activators/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/ultrastructure , Animals , Bezafibrate/administration & dosage , Corpus Striatum/pathology , Diet , Disease Models, Animal , Gliosis/pathology , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/ultrastructure , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Neurons/pathology , Neuroprotective Agents/administration & dosage , Oxidative Stress , PPAR gamma/agonists , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Signal Transduction/drug effects , Signal Transduction/genetics , Survival Rate , Transcription Factors , Transcriptional Activation , Vacuoles/ultrastructure
15.
Neurodegener Dis ; 8(6): 496-503, 2011.
Article in English | MEDLINE | ID: mdl-21757867

ABSTRACT

BACKGROUND: Reduced peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α) gene expression has been observed in striatal cell lines, transgenic mouse models of Huntington's disease (HD), and brain tissue from HD patients. As this protein is a key transcription regulator of the expression of many mitochondrial proteins, these observations strongly support the role of aberrant mitochondrial function in the pathogenesis of HD. The PGC1α protein undergoes posttranslational modifications that affect its transcriptional activity. The N-truncated splice variant of PGC1α (NT-PGC1α) is produced in tissues, but the role of truncated splice variants of PGC1α in HD and in the regulation of mitochondrial gene expression has not been elucidated. OBJECTIVE: To examine the expression and modulation of expression of NT-PGC1α levels in HD. METHODS AND RESULTS: We found that the NT-PGC1α protein, a splice variant of ∼38 kDa, but not full-length PGC1α is severely and consistently altered in human HD brain, human HD myoblasts, mouse HD models, and HD striatal cells. NT-PGC1α levels were significantly upregulated in HD cells and mouse brown fat by physiologically relevant stimuli that are known to upregulate PGC1α gene expression. This resulted in an increase in mitochondrial gene expression and cytochrome c content. CONCLUSION: Our data suggest that NT-PGC1α is an important component of the PGC1α transcriptional network, which plays a significant role in the pathogenesis of HD.


Subject(s)
Heat-Shock Proteins/genetics , Huntington Disease/genetics , Transcription Factors/genetics , Adipose Tissue, Brown/metabolism , Aged , Animals , Blotting, Western , Brain Chemistry/genetics , Brain Chemistry/physiology , Cells, Cultured , Chromatography, High Pressure Liquid , Cold Temperature , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cyclic AMP/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Mitochondria/metabolism , Mutation , Myoblasts/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Isoforms , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/physiology
17.
Neuron ; 67(2): 171-3, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20670824

ABSTRACT

Mutant huntingtin proteolysis mediated by various proteases plays a key role in Huntington's disease (HD) pathogenesis. In this issue of Neuron, Miller et al. have identified 11 proteases, including matrix metalloproteinases (MMPs), that when inhibited reduce huntingtin proteolysis and produce beneficial therapeutic effects. These findings provide new insights into huntingtin proteolysis and its potential as a therapeutic target.

18.
Hum Mol Genet ; 19(16): 3190-205, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20529956

ABSTRACT

We investigated the ability of AMP-activated protein kinase (AMPK) to activate PPARgamma coactivator-1alpha (PGC-1alpha) in the brain, liver and brown adipose tissue (BAT) of the NLS-N171-82Q transgenic mouse model of Huntington's disease (HD). In the striatum of the HD mice, the baseline levels of PGC-1alpha, NRF1, NRF2, Tfam, COX-II, PPARdelta, CREB and ERRalpha mRNA and mitochondrial DNA (mtDNA), were significantly reduced. Administration of the creatine analog beta guanidinopropionic acid (GPA) reduced ATP and PCr levels and increased AMPK mRNA in both the cerebral cortex and striatum. Treatment with GPA significantly increased expression of PGC-1alpha, NRF1, Tfam and downstream genes in the striatum and cerebral cortex of wild-type (WT) mice, but there was no effect on these genes in the HD mice. The striatum of the untreated HD mice showed microvacuolation in the neuropil, as well as gliosis and huntingtin aggregates, which were exacerbated by treatment with GPA. GPA treatment produced a significant increase in mtDNA in the cerebral cortex and striatum of WT mice, but not in HD mice. The HD mice treated with GPA had impaired activation of liver PGC-1alpha and developed hepatic steatosis with accumulation of lipids, degeneration of hepatocytes and impaired activation of gluconeogenesis. The BAT in the HD mice showed vacuolation due to accumulation of neutral lipids, and age-dependent impairment of UCP-1 activation and temperature regulation. Impaired activation of PGC-1alpha, therefore, plays an important role in the behavioral phenotype, metabolic disturbances and pathology of HD, which suggests the possibility that agents that enhance PGC-1alpha function will exert therapeutic benefits in HD patients.


Subject(s)
Disease Models, Animal , Energy Metabolism , Huntington Disease/metabolism , Trans-Activators/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , DNA, Mitochondrial/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression/drug effects , Guanidines/pharmacology , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Immunohistochemistry , Ion Channels/genetics , Ion Channels/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Propionates/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Transcription Factors , Uncoupling Protein 1
19.
Toxicol Appl Pharmacol ; 231(1): 10-6, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18486174

ABSTRACT

Prenatal exposure to low doses of lindane has been shown to affect the ontogeny of xenobiotic metabolizing cytochrome P450s (CYPs), involved in the metabolism and neurobehavioral toxicity of lindane. Attempts were made in the present study to investigate the responsiveness of CYPs in offspring prenatally exposed to lindane (0.25 mg/kg b. wt.; 1/350th of LD(50); p. o. to mother) when challenged with 3-methylcholanthrene (MC) or phenobarbital (PB), inducers of CYP1A and 2B families or a sub-convulsant dose of lindane (30 mg/kg b. wt., p. o.) later in life. Prenatal exposure to lindane was found to produce an increase in the mRNA and protein expression of CYP1A1, 1A2, 2B1, 2B2 isoforms in brain and liver of the offspring at postnatal day 50. The increased expression of the CYPs in the offspring suggests the sensitivity of the CYPs during postnatal development, possibly, to low levels of lindane, which may partition into mother's milk. A higher increase in expression of CYP1A and 2B isoenzymes and their catalytic activity was observed in animals pretreated prenatally with lindane and challenged with MC (30 mg/kg, i. p. x 5 days) or PB (80 mg/kg, i. p. x 5 days) when young at age (approx. 7 weeks) compared to animals exposed to MC or PB alone. Further, challenge of the control and prenatally exposed offspring with a single sub-convulsant dose of lindane resulted in an earlier onset and increased incidence of convulsions in the offspring prenatally exposed to lindane have demonstrated sensitivity of the CYPs in the prenatally exposed offspring. Our data assume significance as the subtle changes in the expression profiles of hepatic and cerebral CYPs in rat offspring during postnatal development could modify the adult response to a later exposure to xenobiotics.


Subject(s)
Brain/enzymology , Cytochrome P-450 Enzyme System/metabolism , Hexachlorocyclohexane/toxicity , Insecticides/toxicity , Liver/enzymology , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain/drug effects , Enzyme Induction/drug effects , Female , Male , Methylcholanthrene/pharmacology , Neurotoxicity Syndromes/psychology , Phenobarbital/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar
20.
Toxicol Sci ; 101(2): 331-40, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17984293

ABSTRACT

Oral administration of low doses (0.0625, 0.125, or 0.25 mg/kg body weight, po, corresponding to 1/1400th, 1/700th, or 1/350th of LD(50), respectively) of lindane, an organochlorine insecticide, to pregnant dams from gestation day 5-21 was found to produce dose-dependent alterations in the ontogenic profile of xenobiotic-metabolizing cytochrome P450s (CYPs) in the brain and liver of offspring. The increase in the cerebral and hepatic mRNA expression of CYP1A1, 1A2, 2B1, 2B2, and 2E1 was also found to be associated with an increase in the catalytic activity of these CYP isoenzymes in the brain and liver of the offspring at different stages during postnatal development. Interestingly, though the levels of CYPs were severalfold lower in brain when compared to the liver, almost equal magnitude of induction in these CYPs in brain have suggested that like in the liver, brain CYPs are responsive to the transplacental induction by environmental chemicals and that the increase is transcriptionally regulated. Moreover, due to its lipophilic nature, lindane may partition in mother's milk leading to further exposure of the offspring during the critical period of neurodevelopment which may explain the increase in CYP mRNA expression and associated catalytic activity especially during the early postnatal period. Interestingly, the increase in mRNA expression of these CYP isoforms was found to persist up to adulthood, suggesting that the low doses of lindane administered to the dams might program the brain and liver of the offspring to persistently express the xenobiotic-metabolizing CYP isoforms. As CYP-dependent metabolism of lindane is involved in its neurobehavioral toxicity, the potential of lindane to imprint the expression of cerebral and hepatic CYPs may help in identifying the role of these enzymes in the developmental neurotoxicity of the pesticide.


Subject(s)
Cerebral Cortex/drug effects , Cytochrome P-450 Enzyme System/biosynthesis , Environmental Pollutants/toxicity , Hexachlorocyclohexane/toxicity , Liver/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Aging/metabolism , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/growth & development , Dose-Response Relationship, Drug , Enzyme Induction , Female , Gestational Age , Isoenzymes , Liver/enzymology , Liver/growth & development , Male , Microsomes/drug effects , Microsomes/enzymology , Pregnancy , Prenatal Exposure Delayed Effects/enzymology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...