Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 294(6): 2060-2073, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30559294

ABSTRACT

Matriptase-2 (MT2) is a type-II transmembrane, trypsin-like serine protease that is predominantly expressed in the liver. It is a key suppressor for the expression of hepatic hepcidin, an iron-regulatory hormone that is induced via the bone morphogenetic protein signaling pathway. A current model predicts that MT2 suppresses hepcidin expression by cleaving multiple components of the induction pathway. MT2 is synthesized as a zymogen that undergoes autocleavage for activation and shedding. However, the biologically active form of MT2 and, importantly, the contributions of different MT2 domains to its function are largely unknown. Here we examined the activities of truncated MT2 that were generated by site-directed mutagenesis or Gibson assembly master mix, and found that the stem region of MT2 determines the specificity and efficacy for substrate cleavage. The transmembrane domain allowed MT2 activation after reaching the plasma membrane, and the cytoplasmic domain facilitated these processes. Further in vivo rescue studies indicated that the entire extracellular and transmembrane domains of MT2 are required to correct the low-hemoglobin, low-serum iron, and high-hepcidin status in MT2-/- mice. Unlike in cell lines, no autocleavage of MT2 was detected in vivo in the liver, implying that MT2 may also function independently of its proteolytic activity. In conjunction with our previous studies implicating the cytoplasmic domain as an intracellular iron sensor, these observations reveal the importance of each MT2 domain for MT2-mediated substrate cleavage and for its biological function.


Subject(s)
Enzyme Precursors/metabolism , Gene Expression Regulation , Hepcidins/biosynthesis , Membrane Proteins/metabolism , Proteolysis , Serine Endopeptidases/metabolism , Animals , Enzyme Precursors/genetics , HEK293 Cells , Hepcidins/genetics , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Serine Endopeptidases/genetics
2.
Mol Endocrinol ; 28(8): 1362-80, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24933249

ABSTRACT

The hypothalamic arcuate nucleus controls many critical homeostatic functions including energy homeostasis, reproduction, and motivated behavior. Although G protein-coupled receptors (GPCRs) are involved in the regulation of these functions, relatively few of the GPCRs have been identified specifically within the arcuate nucleus. Here, using TaqMan low-density arrays we quantified the mRNA expression of nonolfactory GPCRs in mouse arcuate nucleus. An unprecedented number of GPCRs (total of 292) were found to be expressed, of which 183 were known and 109 were orphan GPCRs. The known GPCR genes expressed were classified into several functional clusters including hormone/neurotransmitter, growth factor, angiogenesis and vasoactivity, inflammation and immune system, and lipid messenger receptors. The plethora of orphan genes expressed in the arcuate nucleus were classified into 5 structure-related classes including class A (rhodopsin-like), class B (adhesion), class C (other GPCRs), nonsignaling 7-transmembrane chemokine-binding proteins, and other 7-transmembrane proteins. Therefore, for the first time, we provide a quantitative estimate of the numerous GPCRs expressed in the hypothalamic arcuate nucleus. Finally, as proof of principle, we documented the expression and function of one of these receptor genes, the glucagon-like peptide 1 receptor (Glp1r), which was highly expressed in the arcuate nucleus. Single-cell RT-PCR revealed that Glp1r mRNA was localized in proopiomelanocortin neurons, and using whole-cell recording we found that the glucagon-like peptide 1-selective agonist exendin-4 robustly excited proopiomelanocortin neurons. Thus, the quantitative GPCR data emphasize the complexity of the hypothalamic arcuate nucleus and furthermore provide a valuable resource for future neuroendocrine/endocrine-related experiments.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Receptors, G-Protein-Coupled/genetics , Transcriptome , Animals , Arcuate Nucleus of Hypothalamus/cytology , Cells, Cultured , Female , Gene Expression Profiling , Mice, Inbred C57BL , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
Biochim Biophys Acta ; 1832(12): 2322-31, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055980

ABSTRACT

Oxidative stress and mitochondrial dysfunction are involved in the progression and pathogenesis of multiple sclerosis (MS). MitoQ is a mitochondria-targeted antioxidant that has a neuroprotective role in several mitochondrial and neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Here we sought to determine the possible effects of a systematic administration of MitoQ as a therapy, using an experimental autoimmune encephalomyelitis (EAE) mouse model. We studied the beneficial effects of MitoQ in EAE mice that mimic MS like symptoms by treating EAE mice with MitoQ and pretreated C57BL6 mice with MitoQ plus EAE induction. We found that pretreatment and treatment of EAE mice with MitoQ reduced neurological disabilities associated with EAE. We also found that both pretreatment and treatment of the EAE mice with MitoQ significantly suppressed inflammatory markers of EAE, including the inhibition of inflammatory cytokines and chemokines. MitoQ treatments reduced neuronal cell loss in the spinal cord, a factor underlying motor disability in EAE mice. The neuroprotective role of MitoQ was confirmed by a neuron-glia co-culture system designed to mimic the mechanism of MS and EAE in vitro. We found that axonal inflammation and oxidative stress are associated with impaired behavioral functions in the EAE mouse model and that treatment with MitoQ can exert protective effects on neurons and reduce axonal inflammation and oxidative stress. These protective effects are likely via multiple mechanisms, including the attenuation of the robust immune response. These results suggest that MitoQ may be a new candidate for the treatment of MS.


Subject(s)
Antioxidants/pharmacology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Inflammation/prevention & control , Mitochondria/drug effects , Multiple Sclerosis/prevention & control , Organophosphorus Compounds/pharmacology , Ubiquinone/analogs & derivatives , Animals , Biomarkers/metabolism , Blotting, Western , Coculture Techniques , Cytokines/metabolism , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Ubiquinone/pharmacology
4.
Pharmaceuticals (Basel) ; 6(1): 108-23, 2013.
Article in English | MEDLINE | ID: mdl-23543038

ABSTRACT

Parkinson's disease (PD) is one of the most common neurodegenerative diseases. To date, there is no effective treatment that halts its progression. Increasing evidence indicates that mitochondria play an important role in the development of PD. Hence mitochondria-targeted approaches or agents may have therapeutic promise for treatment of the disease. Neuropeptide CART (cocaine-amphetamine-regulated transcript), a hypothalamus and midbrain enriched neurotransmitter with an antioxidant property, can be found in mitochondria, which is the main source of reactive oxygen species. Systemic administration of CART has been found to ameliorate dopaminergic neuronal loss and improve motor functions in a mouse model of PD. In this article, we summarize recent progress in studies investigating the relationship between CART, dopamine, and the pathophysiology of PD, with a focus on mitochondria-related topics.

5.
J Neurodegener Dis ; 2013: 319898, 2013.
Article in English | MEDLINE | ID: mdl-26316986

ABSTRACT

Dementia is a complex disorder that mostly affects the elderly and represents a significant and growing public health burden in the world. Alzheimer's disease (AD)- associated dementia and dementia with Lewy bodies (DLB) are the most common forms of dementia, in which oxidative stress is significantly involved. Oxidative stress mechanisms may have clinical applications, that is, providing information for potential biomarkers. Thus brain-rich peptides with an antioxidant property, such as CART (cocaine- and amphetamine-regulated transcript), may be promising new markers. This paper summarizes the progress in research regarding oxidative stress in dementia with a focus on potential biomarkers in the cerebrospinal fluid (CSF) in the main forms of dementia. Other central and peripheral biomarkers, especially those considered oxidative stress related, are also discussed. This paper aims to provide information to improve current understanding of the pathogenesis and progression of dementia. It also offers insight into the differential diagnosis of AD and DLB.

6.
Hum Mol Genet ; 21(13): 2973-90, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22492996

ABSTRACT

The purpose of this study was to investigate the protective effects of the mitochondria-targeted antioxidant catalase (MCAT) and lifespan extension in mice that express amyloid beta (Aß). Using immunoblotting and immunostaining analyses, we measured the production of full-length amyloid precursor protein (APP), soluble APPα, C-terminal fragments CTF99 and CTF83, monomeric and oligomeric Aß, Aß deposits and beta site amyloid precursor protein cleaving enzyme 1 (BACE1), in different stages of disease progression in MCAT/AßPP and AßPP mice. Using quantitative reverse transcriptase polymerase chain reaction and immunostaining analyses, we studied the expression of catalase, BACE1, the Alzheimer's disease (AD) markers, synaptophysin, APP, neprilysin, insulin-degrading enzyme and transthyretin in MCAT, AßPP, MCAT/AßPP and wild-type (WT) mice. Using the high pressure liquid chromatography analysis of 8-hydroxy-2-deoxyguanosine, we measured oxidative DNA damage in the cerebral cortical tissues from MCAT, AßPP, MCAT/AßPP and WT mice. We found that the AßPP transgenic mice that carried the human MCAT gene lived 5 months longer than did the AßPP mice. We also found that the overexpression of MCAT in the brain sections from the MCAT/AßPP transgenic mice significantly correlated with a reduction in the levels of full-length APP, CTF99, BACE1, Aß levels (40 and 42), Aß deposits and oxidative DNA damage relative to the brain sections from the AßPP mice. Interestingly, we found significantly increased levels of soluble APPα and CTF83 in the MCAT/AßPP mice, relative to the AßPP mice. These data provide direct evidence that oxidative stress plays a primary role in AD etiopathology and that in MCAT mice express Aß, MCAT prevents abnormal APP processing, reduces Aß levels and enhances Aß-degrading enzymes in mice at different ages, corresponding to different stages of disease progression. These findings indicate that mitochondria-targeted molecules may be an effective therapeutic approach to treat patients with AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/biosynthesis , Catalase/metabolism , Mitochondria/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Animals , Brain/pathology , Catalase/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , DNA Damage/genetics , Disease Models, Animal , Female , Insulysin/biosynthesis , Insulysin/metabolism , Male , Mice , Mice, Transgenic , Neprilysin/biosynthesis , Neuroprotective Agents/metabolism , Oxidative Stress , Prealbumin/biosynthesis , RNA, Messenger/biosynthesis , Random Allocation , Synaptophysin/biosynthesis
7.
ISRN Endocrinol ; 2012: 536905, 2012.
Article in English | MEDLINE | ID: mdl-22474595

ABSTRACT

The prevalence of obesity, which is a heritable trait that arises from the interactions of multiple genes and lifestyle factors, continues to increase worldwide, causing serious health problems and imposing a substantial economic burden on societies. For the past several years, various genetic epidemiological approaches have been utilized to identify genetic loci for obesity. Recent evidence suggests that development of obesity involves hormones and neurotransmitters (such as leptin, cocaine- and amphetamine-regulated transcript (CART), and ghrelin) that regulate appetite and energy expenditure. These hormones act on specific centers in the brain that regulate the sensations of satiety. Mutations in these hormones or their receptors can lead to obesity. Aberrant circadian rhythms and biochemical pathways in peripheral organs or tissues have also been implicated in the pathology of obesity. More interestingly, increasing evidence indicates a potential relation between obesity and central nervous system disorders (such as cognitive deficits). This paper discusses recent advances in the field of genetics of obesity with an emphasis on several established loci that influence obesity. These recently identified loci may hold the promise to substantially improve our insights into the pathophysiology of obesity and open up new therapeutic strategies to combat growing obesity epidemic facing the human population today.

8.
PLoS One ; 7(1): e29343, 2012.
Article in English | MEDLINE | ID: mdl-22235287

ABSTRACT

The multifunctional neuropeptide Cocaine and Amphetamine Regulated Transcript (CART) is secreted from hypothalamus, pituitary, adrenal gland and pancreas. It also can be found in circulatory system. This feature suggests a general role for CART in different cells. In the present study, we demonstrate that CART protects mitochondrial DNA (mtDNA), cellular proteins and lipids against the oxidative action of hydrogen peroxide, a widely used oxidant. Using cis-parinaric acid as a sensitive reporting probe for peroxidation in membranes, and a lipid-soluble azo initiator of peroxyl radicals, 2,2'-azobis(2,4-dimethylvaleronitrile) we found that CART is an antioxidant. Furthermore, we found that CART localized to mitochondria in cultured cells and mouse brain neuronal cells. More importantly, pretreatment with CART by systemic injection protects against a mouse oxidative stress model, which mimics the main features of Parkinson's disease. Given the unique molecular structure and biological features of CART, we conclude that CART is an antioxidant peptide (or antioxidant hormone). We further propose that it may have strong therapeutic properties for human diseases in which oxidative stress is strongly involved such as Parkinson's disease.


Subject(s)
Antioxidants/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Membrane/metabolism , Cell Survival , DNA Damage , DNA, Mitochondrial/genetics , HEK293 Cells , Humans , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Neurons/cytology , Oxidative Stress , Protein Transport , Recombinant Fusion Proteins/metabolism
9.
Hum Mol Genet ; 21(2): 406-20, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21997870

ABSTRACT

The purpose of this study was to investigate the link between mutant huntingtin (Htt) and neuronal damage in relation to mitochondria in Huntington's disease (HD). In an earlier study, we determined the relationship between mutant Htt and mitochondrial dynamics/synaptic viability in HD patients. We found mitochondrial loss, abnormal mitochondrial dynamics and mutant Htt association with mitochondria in HD patients. In the current study, we sought to expand on our previous findings and further elucidate the relationship between mutant Htt and mitochondrial and synaptic deficiencies. We hypothesized that mutant Htt, in association with mitochondria, alters mitochondrial dynamics, leading to mitochondrial fragmentation and defective axonal transport of mitochondria in HD neurons. In this study, using postmortem HD brains and primary neurons from transgenic BACHD mice, we identified mutant Htt interaction with the mitochondrial protein Drp1 and factors that cause abnormal mitochondrial dynamics, including GTPase Drp1 enzymatic activity. Further, using primary neurons from BACHD mice, for the first time, we studied axonal transport of mitochondria and synaptic degeneration. We also investigated the effect of mutant Htt aggregates and oligomers in synaptic and mitochondrial deficiencies in postmortem HD brains and primary neurons from BACHD mice. We found that mutant Htt interacts with Drp1, elevates GTPase Drp1 enzymatic activity, increases abnormal mitochondrial dynamics and results in defective anterograde mitochondrial movement and synaptic deficiencies. These observations support our hypothesis and provide data that can be utilized to develop therapeutic targets that are capable of inhibiting mutant Htt interaction with Drp1, decreasing mitochondrial fragmentation, enhancing axonal transport of mitochondria and protecting synapses from toxic insults caused by mutant Htt.


Subject(s)
Axons , GTP Phosphohydrolases/metabolism , Huntington Disease/pathology , Microtubule-Associated Proteins/metabolism , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Synapses/pathology , Animals , Dynamins , Humans , Huntingtin Protein , Huntington Disease/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Binding
10.
Biochim Biophys Acta ; 1822(5): 639-49, 2012 May.
Article in English | MEDLINE | ID: mdl-22037588

ABSTRACT

Synaptic pathology and mitochondrial oxidative damage are early events in Alzheimer's disease (AD) progression. Loss of synapses and synaptic damage are the best correlates of cognitive deficits found in AD patients. Recent research on amyloid beta (Aß) and mitochondria in AD revealed that Aß accumulates in synapses and synaptic mitochondria, leading to abnormal mitochondrial dynamics and synaptic degeneration in AD neurons. Further, recent studies using live-cell imaging and primary neurons from amyloid beta precursor protein (AßPP) transgenic mice revealed reduced mitochondrial mass, defective axonal transport of mitochondria and synaptic degeneration, indicating that Aß is responsible for mitochondrial and synaptic deficiencies. Tremendous progress has been made in studying antioxidant approaches in mouse models of AD and clinical trials of AD patients. This article highlights the recent developments made in Aß-induced abnormal mitochondrial dynamics, defective mitochondrial biogenesis, impaired axonal transport and synaptic deficiencies in AD. This article also focuses on mitochondrial approaches in treating AD, and also discusses latest research on mitochondria-targeted antioxidants in AD. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.


Subject(s)
Alzheimer Disease/physiopathology , Antioxidants/therapeutic use , Mitochondria/drug effects , Synapses/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Antioxidants/pharmacology , Humans , Mice , Mice, Transgenic , Mitochondria/physiology
11.
Biochim Biophys Acta ; 1822(2): 111-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22056405

ABSTRACT

The purpose of this study was to determine the relationship between mitochondrial DNA (mtDNA) deletions, mtDNA content and aging in rhesus monkeys. Using 2 sets of specific primers, we amplified an 8 kb mtDNA fragment covering a common 5.7 kb deletion and the entire 16.5 kb mitochondrial genome in the brain and buffy-coats of young and aged monkeys. We studied a total of 66 DNA samples: 39 were prepared from a buffy-coat and 27 were prepared from occipital cortex tissues. The mtDNA data were assessed using a permutation test to identify differences in mtDNA, in the different monkey groups. Using real-time RT-PCR strategy, we also assessed both mtDNA and nuclear DNA levels for young, aged and male and female monkeys. We found a 5.7 kb mtDNA deletion in 81.8% (54 of 66) of the total tested samples. In the young group of buffy-coat DNA, we found 5.7 kb deletions in 7 of 17 (41%), and in the aged group, we found 5.7 kb deletions in 12 of 22 (54%), suggesting that the prevalence of mtDNA deletions is related to age. We found decreased mRNA levels of mtDNA in aged monkeys relative to young monkeys. The increases in mtDNA deletions and mtDNA levels in aged rhesus monkeys suggest that damaged DNA accumulates as rhesus monkeys age and these altered mtDNA changes may have physiological relevance to compensate decreased mitochondrial function.


Subject(s)
Aging/genetics , DNA Damage/genetics , DNA, Mitochondrial/genetics , Genome, Mitochondrial , Macaca mulatta/genetics , Mitochondria/genetics , Animals , Blood Buffy Coat/metabolism , Brain/metabolism , Female , Gene Deletion , Macaca mulatta/blood , Male
12.
Hum Mol Genet ; 20(23): 4515-29, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21873260

ABSTRACT

Increasing evidence suggests that the accumulation of amyloid beta (Aß) in synapses and synaptic mitochondria causes synaptic mitochondrial failure and synaptic degeneration in Alzheimer's disease (AD). The purpose of this study was to better understand the effects of Aß in mitochondrial activity and synaptic alterations in neurons from a mouse model of AD. Using primary neurons from a well-characterized Aß precursor protein transgenic (AßPP) mouse model (Tg2576 mouse line), for the first time, we studied mitochondrial activity, including axonal transport of mitochondria, mitochondrial dynamics, morphology and function. Further, we also studied the nature of Aß-induced synaptic alterations, and cell death in primary neurons from Tg2576 mice, and we sought to determine whether the mitochondria-targeted antioxidant SS31 could mitigate the effects of oligomeric Aß. We found significantly decreased anterograde mitochondrial movement, increased mitochondrial fission and decreased fusion, abnormal mitochondrial and synaptic proteins and defective mitochondrial function in primary neurons from AßPP mice compared with wild-type (WT) neurons. Transmission electron microscopy revealed a large number of small mitochondria and structurally damaged mitochondria, with broken cristae in AßPP primary neurons. We also found an increased accumulation of oligomeric Aß and increased apoptotic neuronal death in the primary neurons from the AßPP mice relative to the WT neurons. Our results revealed an accumulation of intraneuronal oligomeric Aß, leading to mitochondrial and synaptic deficiencies, and ultimately causing neurodegeneration in AßPP cultures. However, we found that the mitochondria-targeted antioxidant SS31 restored mitochondrial transport and synaptic viability, and decreased the percentage of defective mitochondria, indicating that SS31 protects mitochondria and synapses from Aß toxicity.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Axonal Transport , Mitochondria/metabolism , Mitochondria/pathology , Synapses/pathology , Adenosine Triphosphate/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Animals , Apoptosis/drug effects , Axonal Transport/drug effects , Blotting, Western , Cells, Cultured , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Disease Models, Animal , Electron Transport Complex IV/metabolism , Gene Expression Regulation/drug effects , Hydrogen Peroxide/metabolism , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Molecular Weight , Neurites/drug effects , Neurites/metabolism , Neurites/pathology , Oligopeptides/pharmacology , Peroxiredoxins/metabolism , Protein Structure, Quaternary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synapses/drug effects , Synapses/metabolism
13.
Biochim Biophys Acta ; 1812(11): 1359-70, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21871956

ABSTRACT

Alzheimer's disease (AD) is an age-related progressive neurodegenerative disease affecting thousands of people in the world and effective treatment is still not available. Over two decades of intense research using AD postmortem brains, transgenic mouse and cell models of amyloid precursor protein and tau revealed that amyloid beta (Aß) and hyperphosphorylated tau are synergistically involved in triggering disease progression. Accumulating evidence also revealed that aging and amyloid beta-induced oxidative DNA damage and mitochondrial dysfunction initiate and contributes to the development and progression of the disease. The purpose of this article is to summarize the latest progress in aging and AD, with a special emphasis on the mitochondria, oxidative DNA damage including methods of its measurement. It also discusses the therapeutic approaches against oxidative DNA damage and treatment strategies in AD.


Subject(s)
Aging/physiology , Alzheimer Disease/etiology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/adverse effects , DNA Damage , Mitochondria/pathology , Alzheimer Disease/pathology , Animals , Humans , Mice , Oxidation-Reduction , Oxidative Stress
14.
Neurosci Lett ; 500(3): 212-5, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21741442

ABSTRACT

The chronic accumulation of amyloid beta (Aß) peptides is thought to underlie much of the pathology of Alzheimer's disease (AD), and transgenic mice overexpressing Aß show both behavioral defects and impairments in hippocampal synaptic transmission. In the present study, we examined excitatory transmission at the Schaffer collateral synapse in acute hippocampal slices from APP(Swe)/PS-1(A246E) transgenic mice to determine whether the synaptic impairment in these mice is due to a reduction in the activity-independent synaptic gain, or to a change in the activity-dependent synaptic dynamics. We observed a strong reduction in synaptic transmission in slices from APP(Swe)/PS-1(A246E) mice compared to those from their wildtype littermates. However, there was no resolvable change in the synaptic dynamics observed in response to either simple or complex stimulus trains. We conclude that the chronic accumulation of Aß impairs synaptic transmission through a reduction in the synaptic gain, while preserving the synaptic dynamics.


Subject(s)
Alzheimer Disease/physiopathology , Synapses/physiology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Excitatory Postsynaptic Potentials , Hippocampus/physiopathology , In Vitro Techniques , Mice , Mice, Transgenic , Presenilin-1/genetics , Synaptic Transmission
15.
Depress Res Treat ; 2011: 762139, 2011.
Article in English | MEDLINE | ID: mdl-21785720

ABSTRACT

Depression is one of the most prevalent and debilitating public health concerns. Although no single cause of depression has been identified, it appears that interaction among genetic, epigenetic, biochemical, environmental, and psychosocial factors may explain its etiology. Further, only a fraction of depressed patients show full remission while using current antidepressants. Therefore, identifying common pathways of the disorder and using that knowledge to develop more effective pharmacological treatments are two primary targets of research in this field. Brain-enriched neurotransmitter CART (cocaine- and amphetamine-regulated transcript) has multiple functions related to emotions. It is a potential neurotrophic factor and is involved in the regulation of hypothalamic-pituitary-adrenal axis and stress response as well as in energy homeostasis. CART is also highly expressed in limbic system, which is considered to have an important role in regulating mood. Notably, adolescents carrying a missense mutation in the CART gene exhibit increased depression and anxiety. Hence, CART peptide may be a novel promising antidepressant agent. In this paper, we summarize recent progress in depression and CART. In particular, we emphasize a new antidepressant function for CART.

16.
Int J Environ Res Public Health ; 8(1): 203-21, 2011 01.
Article in English | MEDLINE | ID: mdl-21318024

ABSTRACT

The purpose of this study was to determine the neurotoxicity of two commonly used herbicides: picloram and triclopyr and the neuroprotective effects of the mitochondria-targeted antioxidant, SS31. Using mouse neuroblastoma (N2a) cells and primary neurons from C57BL/6 mice, we investigated the toxicity of these herbicides, and protective effects of SS1 peptide against picloram and triclopyr toxicity. We measured total RNA content, cell viability and mRNA expression of peroxiredoxins, neuroprotective genes, mitochondrial-encoded electron transport chain (ETC) genes in N2a cells treated with herbicides and SS31. Using primary neurons from C57BL/6 mice, neuronal survival was studied in neurons treated with herbicides, in neurons pretreated with SS31 plus treated with herbicides, neurons treated with SS31 alone, and untreated neurons. Significantly decreased total RNA content, and cell viability in N2a cells treated with picloram and triclopyr were found compared to untreated N2a cells. Decreased mRNA expression of neuroprotective genes, and ETC genes in cells treated with herbicides was found compared to untreated cells. Decreased mRNA expression of peroxiredoxins 1-6 in N2a cells treated with picloram was found, suggesting that picloram affects the antioxidant enzymes in N2a cells. Immunofluorescence analysis of primary neurons revealed that decreased neuronal branching and degenerating neurons in neurons treated with picloram and triclopyr. However, neurons pretreated with SS31 prevented degenerative process caused by herbicides. Based on these results, we propose that herbicides--picloram and triclopyr appear to damage neurons, and the SS31 peptide appears to protect neurons from herbicide toxicity.


Subject(s)
Antioxidants/physiology , Glycolates/toxicity , Herbicides/toxicity , Neurons/drug effects , Oligopeptides/physiology , Picloram/toxicity , Animals , Antioxidants/administration & dosage , Cell Line, Tumor , Cell Survival , Electron Transport Chain Complex Proteins/biosynthesis , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , Mice , Mice, Inbred C57BL , Neuroblastoma , Neurons/pathology , Neurons/physiology , Oligopeptides/administration & dosage , Oxidative Stress , Peroxiredoxins/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Receptors, N-Methyl-D-Aspartate/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/biosynthesis , Transcription Factors , Transcription, Genetic
17.
Hum Mol Genet ; 20(7): 1438-55, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21257639

ABSTRACT

The purpose of our study was to determine the relationship between mutant huntingtin (Htt) and mitochondrial dynamics in the progression of Huntington's disease (HD). We measured the mRNA levels of electron transport chain genes, and mitochondrial structural genes, Drp1 (dynamin-related protein 1), Fis1 (fission 1), Mfn1 (mitofusin 1), Mfn2 (mitofusin 2), Opa1 (optric atrophy 1), Tomm40 (translocase of outermembrane 40) and CypD (cyclophilin D) in grade III and grade IV HD patients and controls. The mutant Htt oligomers and the mitochondrial structural proteins were quantified in the striatum and frontal cortex of HD patients. Changes in expressions of the electron transport chain genes were found in HD patients and may represent a compensatory response to mitochondrial damage caused by mutant Htt. Increased expression of Drp1 and Fis1 and decreased expression of Mfn1, Mfn2, Opa1 and Tomm40 were found in HD patients relative to the controls. CypD was upregulated in HD patients, and this upregulation increased as HD progressed. Significantly increased immunoreactivity of 8-hydroxy-guanosine was found in the cortical specimens from stage III and IV HD patients relative to controls, suggesting increased oxidative DNA damage in HD patients. In contrast, significantly decreased immunoreactivities of cytochrome oxidase 1 and cytochrome b were found in HD patients relative to controls, indicating a loss of mitochondrial function in HD patients. Immunoblotting analysis revealed 15, 25 and 50 kDa mutant Htt oligomers in the brain specimens of HD patients. All oligomeric forms of mutant Htt were significantly increased in the cortical tissues of HD patients, and mutant Htt oligomers were found in the nucleus and in mitochondria. The increase in Drp1, Fis1 and CypD and the decrease in Mfn1 and Mfn2 may be responsible for abnormal mitochondrial dynamics that we found in the cortex of HD patients, and may contribute to neuronal damage in HD patients. The presence of mutant Htt oligomers in the nucleus of HD neurons and in mitochondria may disrupt neuronal functions. Based on these findings, we propose that mutant Htt in association with mitochondria imbalance and mitochondrial dynamics impairs axonal transport of mitochondria, decreases mitochondrial function and damages neurons in affected brain regions of HD patients.


Subject(s)
Axons/metabolism , Frontal Lobe/metabolism , Huntington Disease/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Multimerization , Axons/pathology , Biological Transport/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , DNA Damage/genetics , Electron Transport Chain Complex Proteins/biosynthesis , Electron Transport Chain Complex Proteins/genetics , Female , Frontal Lobe/pathology , Gene Expression Regulation/genetics , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Male , Mitochondria/genetics , Mitochondria/pathology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
18.
Brain Res Rev ; 67(1-2): 103-18, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21145355

ABSTRACT

The purpose of this article is to review the recent developments of abnormal mitochondrial dynamics, mitochondrial fragmentation, and neuronal damage in neurodegenerative diseases, including Alzheimer's, Parkinson's, Huntington's, and amyotrophic lateral sclerosis. The GTPase family of proteins, including fission proteins, dynamin related protein 1 (Drp1), mitochondrial fission 1 (Fis1), and fusion proteins (Mfn1, Mfn2 and Opa1) are essential to maintain mitochondrial fission and fusion balance, and to provide necessary adenosine triphosphate to neurons. Among these, Drp1 is involved in several important aspects of mitochondria, including shape, size, distribution, remodeling, and maintenance of mitochondria in mammalian cells. In addition, recent advancements in molecular, cellular, electron microscopy, and confocal imaging studies revealed that Drp1 is associated with several cellular functions, including mitochondrial and peroxisomal fragmentation, phosphorylation, SUMOylation, ubiquitination, and cell death. In the last two decades, tremendous progress has been made in researching mitochondrial dynamics, in yeast, worms, and mammalian cells; and this research has provided evidence linking Drp1 to neurodegenerative diseases. Researchers in the neurodegenerative disease field are beginning to recognize the possible involvement of Drp1 in causing mitochondrial fragmentation and abnormal mitochondrial dynamics in neurodegenerative diseases. This article summarizes research findings relating Drp1 to mitochondrial fission and fusion, in yeast, worms, and mammals. Based on findings from the Reddy laboratory and others', we propose that mutant proteins of neurodegenerative diseases, including AD, PD, HD, and ALS, interact with Drp1, activate mitochondrial fission machinery, fragment mitochondria excessively, and impair mitochondrial transport and mitochondrial dynamics, ultimately causing mitochondrial dysfunction and neuronal damage.


Subject(s)
GTP Phosphohydrolases/physiology , Microtubule-Associated Proteins/physiology , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Animals , Cell Respiration/genetics , Cell Respiration/physiology , Dynamins , GTP Phosphohydrolases/genetics , Humans , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Models, Animal , Mutation , Neurodegenerative Diseases/genetics
19.
J Alzheimers Dis ; 20 Suppl 2: S609-31, 2010.
Article in English | MEDLINE | ID: mdl-20463406

ABSTRACT

The purpose of our study was to investigate the effects of the mitochondria-targeted antioxidants, MitoQ and SS31, and the anti-aging agent resveratrol on neurons from a mouse model (Tg2576 line) of Alzheimer's disease (AD) and on mouse neuroblastoma (N2a) cells incubated with the amyloid-beta (Abeta) peptide. Using electron and confocal microscopy, gene expression analysis, and biochemical methods, we studied mitochondrial structure and function and neurite outgrowth in N2a cells treated with MitoQ, SS31, and resveratrol, and then incubated with Abeta. In N2a cells only incubated with the Abeta, we found increased expressions of mitochondrial fission genes and decreased expression of fusion genes and also decreased expression of peroxiredoxins. Electron microscopy of the N2a cells incubated with Abeta revealed a significantly increased number of mitochondria, indicating that Abeta fragments mitochondria. Biochemical analysis revealed that function is defective in mitochondria. Neurite outgrowth was significantly decreased in Abeta-incubated N2a cells, indicating that Abeta affects neurite outgrowth. However, in N2a cells treated with MitoQ, SS31, and resveratrol, and then incubated with Abeta, abnormal expression of peroxiredoxins and mitochondrial structural genes were prevented and mitochondrial function was normal; intact mitochondria were present and neurite outgrowth was significantly increased. In primary neurons from amyloid-beta precursor protein transgenic mice that were treated with MitoQ and SS31, neurite outgrowth was significantly increased and cyclophilin D expression was significantly decreased. These findings suggest that MitoQ and SS31 prevent Abeta toxicity, which would warrant the study of MitoQ and SS31 as potential drugs to treat patients with AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Antioxidants/pharmacology , Gene Expression Regulation/drug effects , Mitochondria/drug effects , Neurons/drug effects , Peptide Fragments/toxicity , Adenosine Triphosphate/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor , Analysis of Variance , Animals , Cell Line, Transformed/ultrastructure , Cell Survival/drug effects , Disease Models, Animal , Drug Interactions , Electron Transport Complex IV/metabolism , Humans , Hydrogen Peroxide/metabolism , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , RNA, Messenger/metabolism
20.
J Alzheimers Dis ; 20 Suppl 2: S499-512, 2010.
Article in English | MEDLINE | ID: mdl-20413847

ABSTRACT

This article reviews the role of amyloid-beta (Abeta) and mitochondria in synaptic damage and cognitive decline found in patients with Alzheimer's disease (AD). Recent molecular, cellular, animal model, and postmortem brain studies have revealed that Abeta and mitochondrial abnormalities are key factors that cause synaptic damage and cognitive decline in AD. Abeta is reported to accumulate in subcellular compartments and to impair the normal function of neurons in AD patients. Further, recent studies using biochemical methods and electron microscopy have revealed that the accumulation of Abeta at nerve terminals affect synaptic activities, including the release of neurotransmitters and synaptic vesicles. Recent studies of the relationship between mitochondria and Abeta in AD patients suggest that in mitochondria, structural changes caused by Abeta result in increased mitochondrial fragmentation, decreased mitochondrial fusion, mitochondrial dysfunction, and synaptic damage. This paper discusses the latest research on Abeta, mitochondria, age-dependent factors of AD in the brain, and synaptic damage in AD. This paper also briefly discusses potential mitochondrial therapeutics in the treatment of patients with AD.


Subject(s)
Aging/pathology , Alzheimer Disease , Amyloid beta-Peptides/metabolism , Cognition Disorders/etiology , Mitochondria/pathology , Synapses/pathology , Aging/metabolism , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Humans , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL