Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Am J Hum Genet ; 111(3): 473-486, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38354736

RESUMO

Disease-associated variants identified from genome-wide association studies (GWASs) frequently map to non-coding areas of the genome such as introns and intergenic regions. An exclusive reliance on gene-agnostic methods of genomic investigation could limit the identification of relevant genes associated with polygenic diseases such as Alzheimer disease (AD). To overcome such potential restriction, we developed a gene-constrained analytical method that considers only moderate- and high-risk variants that affect gene coding sequences. We report here the application of this approach to publicly available datasets containing 181,388 individuals without and with AD and the resulting identification of 660 genes potentially linked to the higher AD prevalence among Africans/African Americans. By integration with transcriptome analysis of 23 brain regions from 2,728 AD case-control samples, we concentrated on nine genes that potentially enhance the risk of AD: AACS, GNB5, GNS, HIPK3, MED13, SHC2, SLC22A5, VPS35, and ZNF398. GNB5, the fifth member of the heterotrimeric G protein beta family encoding Gß5, is primarily expressed in neurons and is essential for normal neuronal development in mouse brain. Homozygous or compound heterozygous loss of function of GNB5 in humans has previously been associated with a syndrome of developmental delay, cognitive impairment, and cardiac arrhythmia. In validation experiments, we confirmed that Gnb5 heterozygosity enhanced the formation of both amyloid plaques and neurofibrillary tangles in the brains of AD model mice. These results suggest that gene-constrained analysis can complement the power of GWASs in the identification of AD-associated genes and may be more broadly applicable to other polygenic diseases.


Assuntos
Doença de Alzheimer , Subunidades beta da Proteína de Ligação ao GTP , Camundongos , Humanos , Animais , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Estudo de Associação Genômica Ampla , Emaranhados Neurofibrilares/metabolismo , Fenótipo , Genômica , Peptídeos beta-Amiloides/genética , Encéfalo/metabolismo , Membro 5 da Família 22 de Carreadores de Soluto/genética , Membro 5 da Família 22 de Carreadores de Soluto/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo
2.
Heliyon ; 9(10): e20384, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37780758

RESUMO

Oligodendrocytes (OLs) generate lipid-rich myelin membranes that wrap axons to enable efficient transmission of electrical impulses. Using a RIT1 knockout mouse model and in situ high-resolution matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) coupled with MS-based lipidomic analysis to determine the contribution of RIT1 to lipid homeostasis. Here, we report that RIT1 loss is associated with altered lipid levels in the central nervous system (CNS), including myelin-associated lipids within the corpus callosum (CC). Perturbed lipid metabolism was correlated with reduced numbers of OLs, but increased numbers of GFAP+ glia, in the CC, but not in grey matter. This was accompanied by reduced myelin protein expression and axonal conduction deficits. Behavioral analyses revealed significant changes in voluntary locomotor activity and anxiety-like behavior in RIT1KO mice. Together, these data reveal an unexpected role for RIT1 in the regulation of cerebral lipid metabolism, which coincide with altered white matter tract oligodendrocyte levels, reduced axonal conduction velocity, and behavioral abnormalities in the CNS.

3.
JCI Insight ; 8(13)2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37219953

RESUMO

Mechanical, thermal, and chemical pain sensation is conveyed by primary nociceptors, a subset of sensory afferent neurons. The intracellular regulation of the primary nociceptive signal is an area of active study. We report here the discovery of a Gß5-dependent regulatory pathway within mechanical nociceptors that restrains antinociceptive input from metabotropic GABA-B receptors. In mice with conditional knockout (cKO) of the gene that encodes Gß5 (Gnb5) targeted to peripheral sensory neurons, we demonstrate the impairment of mechanical, thermal, and chemical nociception. We further report the specific loss of mechanical nociception in Rgs7-Cre+/- Gnb5fl/fl mice but not in Rgs9-Cre+/- Gnb5fl/fl mice, suggesting that Gß5 might specifically regulate mechanical pain in regulator of G protein signaling 7-positive (Rgs7+) cells. Additionally, Gß5-dependent and Rgs7-associated mechanical nociception is dependent upon GABA-B receptor signaling since both were abolished by treatment with a GABA-B receptor antagonist and since cKO of Gß5 from sensory cells or from Rgs7+ cells potentiated the analgesic effects of GABA-B agonists. Following activation by the G protein-coupled receptor Mrgprd agonist ß-alanine, enhanced sensitivity to inhibition by baclofen was observed in primary cultures of Rgs7+ sensory neurons harvested from Rgs7-Cre+/- Gnb5fl/fl mice. Taken together, these results suggest that the targeted inhibition of Gß5 function in Rgs7+ sensory neurons might provide specific relief for mechanical allodynia, including that contributing to chronic neuropathic pain, without reliance on exogenous opioids.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP , Proteínas RGS , Animais , Camundongos , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Nociceptividade , Transdução de Sinais/fisiologia , Dor , Proteínas RGS/genética , Proteínas RGS/metabolismo
4.
Am J Physiol Endocrinol Metab ; 320(2): E270-E280, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33166186

RESUMO

The G-protein subunits Gqα and G11α (Gq/11α) couple receptors to phospholipase C, leading to increased intracellular calcium. In this study we investigated the consequences of Gq/11α deficiency in the dorsomedial hypothalamus (DMH), a critical site for the control of energy homeostasis. Mice with DMH-specific deletion of Gq/11α (DMHGq/11KO) were generated by stereotaxic injection of adeno-associated virus (AAV)-Cre-green fluorescent protein (GFP) into the DMH of Gqαflox/flox:G11α-/- mice. Compared with control mice that received DMH injection of AAV-GFP, DMHGq/11KO mice developed obesity associated with reduced energy expenditure without significant changes in food intake or physical activity. DMHGq/11KO mice showed no defects in the ability of the melanocortin agonist melanotan II to acutely stimulate energy expenditure or to inhibit food intake. At room temperature (22°C), DMHGq/11KO mice showed reduced sympathetic nervous system activity in brown adipose tissue (BAT) and heart, accompanied with decreased basal BAT uncoupling protein 1 (Ucp1) gene expression and lower heart rates. These mice were cold intolerant when acutely exposed to cold (6°C for 5 h) and had decreased cold-stimulated BAT Ucp1 gene expression. DMHGq/11KO mice also failed to adapt to gradually declining ambient temperatures and to develop adipocyte browning in inguinal white adipose tissue although their BAT Ucp1 was proportionally stimulated. Consistent with impaired cold-induced thermogenesis, the onset of obesity in DMHGq/11KO mice was significantly delayed when housed under thermoneutral conditions (30°C). Thus our results show that Gqα and G11α in the DMH are required for the control of energy homeostasis by stimulating energy expenditure and thermoregulation.NEW & NOTEWORTHY This paper demonstrates that signaling within the dorsomedial hypothalamus via the G proteins Gqα and G11α, which couple cell surface receptors to the stimulation of phospholipase C, is critical for regulation of energy expenditure, thermoregulation by brown adipose tissue and the induction of white adipose tissue browning.


Assuntos
Doenças do Sistema Nervoso Autônomo/genética , Metabolismo Energético/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Hipotálamo/metabolismo , Obesidade/genética , Animais , Doenças do Sistema Nervoso Autônomo/metabolismo , Doenças do Sistema Nervoso Autônomo/fisiopatologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Obesidade/fisiopatologia , Especificidade de Órgãos/genética , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiopatologia
5.
Commun Biol ; 2: 338, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31531399

RESUMO

Protein-protein interaction (PPI) networks are known to be valuable targets for therapeutic intervention; yet the development of PPI modulators as next-generation drugs to target specific vertices, edges, and hubs has been impeded by the lack of structural information of many of the proteins and complexes involved. Building on recent advancements in cross-linking mass spectrometry (XL-MS), we describe an effective approach to obtain relevant structural data on R7BP, a master regulator of itch sensation, and its interfaces with other proteins in its network. This approach integrates XL-MS with a variety of modeling techniques to successfully develop antibody inhibitors of the R7BP and RGS7/Gß5 duplex interaction. Binding and inhibitory efficiency are studied by surface plasmon resonance spectroscopy and through an R7BP-derived dominant negative construct. This approach may have broader applications as a tool to facilitate the development of PPI modulators in the absence of crystal structures or when structural information is limited.


Assuntos
Desenho de Fármacos , Modelos Moleculares , Proteínas RGS/antagonistas & inibidores , Proteínas RGS/química , Sequência de Aminoácidos , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Sítios de Ligação , Descoberta de Drogas , Humanos , Ligantes , Conformação Molecular , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
6.
J Genet ; 97(3): 649-664, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30027901

RESUMO

Huntington's disease (HD) is caused due to an abnormal expansion of polyglutamine repeats in the first exon of huntingtin gene. The mutation in huntingtin causes abnormalities in the functioning of protein, leading to deleterious effects ultimately to the demise of specific neuronal cells.The disease is inherited in an autosomal dominant manner and leads to a plethora of neuropsychiatric behaviour and neuronal cell death mainly in striatal and cortical regions of the brain, eventually leading to death of the individual. The discovery of the mutant gene led to a surge in molecular diagnostics of the disease and in making different transgenic models in different organisms to understand the function of wild-type and mutant proteins. Despite difficult challenges, there has been a significant increase in understanding the functioning of the protein in normal and other gain-of-function interactions in mutant form. However, there have been no significant improvements in treatments of the patients suffering from this ailment and most of the treatment is still symptomatic. HD warrants more attention towards better understanding and treatment as more advancement in molecular diagnostics and therapeutic interventions are available. Several different transgenic models are available in different organisms, ranging from fruit flies to primate monkeys, for studies on understanding the pathogenicity of the mutant gene. It is the right time to assess the advancement in the field and try new strategies for neuroprotection using key pathways as target. The present review highlights the key ingredients of pathology in the HD and discusses important studies for drug trials and future goals for therapeutic interventions.


Assuntos
Doença de Huntington/patologia , Animais , Humanos , Doença de Huntington/epidemiologia , Doença de Huntington/fisiopatologia , Doença de Huntington/terapia , Mutação/genética , Neuroproteção , Prevalência
7.
Pain ; 158(5): 931-944, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28134655

RESUMO

Itch is a protective sensation producing a desire to scratch. Pathologic itch can be a chronic symptom of illnesses such as uremia, cholestatic liver disease, neuropathies and dermatitis, however current therapeutic options are limited. Many types of cell surface receptors, including those present on cells in the skin, on sensory neurons and on neurons in the spinal cord, have been implicated in itch signaling. The role of G protein signaling in the regulation of pruriception is poorly understood. We identify here 2 G protein signaling components whose mutation impairs itch sensation. R7bp (a.k.a. Rgs7bp) is a palmitoylated membrane anchoring protein expressed in neurons that facilitates Gαi/o -directed GTPase activating protein activity mediated by the Gß5/R7-RGS complex. Knockout of R7bp diminishes scratching responses to multiple cutaneously applied and intrathecally-administered pruritogens in mice. Knock-in to mice of a GTPase activating protein-insensitive mutant of Gαo (Gnao1 G184S/+) produces a similar pruriceptive phenotype. The pruriceptive defect in R7bp knockout mice was rescued in double knockout mice also lacking Oprk1, encoding the G protein-coupled kappa-opioid receptor whose activation is known to inhibit itch sensation. In a model of atopic dermatitis (eczema), R7bp knockout mice showed diminished scratching behavior and enhanced sensitivity to kappa opioid agonists. Taken together, our results indicate that R7bp is a key regulator of itch sensation and suggest the potential targeting of R7bp-dependent GTPase activating protein activity as a novel therapeutic strategy for pathological itch.


Assuntos
Nociceptividade/fisiologia , Prurido/genética , Prurido/metabolismo , Proteínas RGS/metabolismo , Sensação/genética , Animais , Cânfora/análogos & derivados , Cânfora/toxicidade , Células Cultivadas , Cromonas/toxicidade , Endotelina-1/toxicidade , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Gânglios Espinais/patologia , Peptídeo Liberador de Gastrina/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Peptídeo Natriurético Encefálico/toxicidade , Limiar da Dor/fisiologia , Prurido/induzido quimicamente , Desempenho Psicomotor/fisiologia , Proteínas RGS/genética , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia
8.
Bioengineered ; 7(3): 166-74, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27340770

RESUMO

CRISPR (Clustered Regularly-Interspaced Short Palindromic Repeats)-Cas9 (CRISPR associated protein 9) has rapidly become the most promising genome editing tool with great potential to revolutionize medicine. Through guidance of a 20 nucleotide RNA (gRNA), CRISPR-Cas9 finds and cuts target protospacer DNA precisely 3 base pairs upstream of a PAM (Protospacer Adjacent Motif). The broken DNA ends are repaired by either NHEJ (Non-Homologous End Joining) resulting in small indels, or by HDR (Homology Directed Repair) for precise gene or nucleotide replacement. Theoretically, CRISPR-Cas9 could be used to modify any genomic sequences, thereby providing a simple, easy, and cost effective means of genome wide gene editing. However, the off-target activity of CRISPR-Cas9 that cuts DNA sites with imperfect matches with gRNA have been of significant concern because clinical applications require 100% accuracy. Additionally, CRISPR-Cas9 has unpredictable efficiency among different DNA target sites and the PAM requirements greatly restrict its genome editing frequency. A large number of efforts have been made to address these impeding issues, but much more is needed to fully realize the medical potential of CRISPR-Cas9. In this article, we summarize the existing problems and current advances of the CRISPR-Cas9 technology and provide perspectives for the ultimate perfection of Cas9-mediated genome editing.


Assuntos
Proteínas de Bactérias/genética , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Endonucleases/genética , Edição de Genes/métodos , Genoma , RNA Guia de Cinetoplastídeos/genética , Animais , Proteínas de Bactérias/metabolismo , Proteína 9 Associada à CRISPR , DNA/química , DNA/genética , DNA/metabolismo , Reparo do DNA por Junção de Extremidades , Endonucleases/metabolismo , Humanos , Modelos Moleculares , Motivos de Nucleotídeos , RNA Guia de Cinetoplastídeos/metabolismo , Reparo de DNA por Recombinação
9.
J Clin Invest ; 126(1): 40-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26595811

RESUMO

Activation of brain melanocortin 4 receptors (MC4Rs) leads to reduced food intake, increased energy expenditure, increased insulin sensitivity, and reduced linear growth. MC4R effects on energy expenditure and glucose metabolism are primarily mediated by the G protein G(s)α in brain regions outside of the paraventricular nucleus of the hypothalamus (PVN). However, the G protein(s) that is involved in MC4R-mediated suppression of food intake and linear growth, which are believed to be regulated primarily though action in the PVN, is unknown. Here, we show that PVN-specific loss of G(q)α and G11α, which stimulate PLC, leads to severe hyperphagic obesity, increased linear growth, and inactivation of the hypothalamic-pituitary-adrenal axis, without affecting energy expenditure or glucose metabolism. Moreover, we demonstrate that the ability of an MC4R agonist delivered to PVN to inhibit food intake is lost in mice lacking G(q/11)α in the PVN but not in animals deficient for G(s)α. The blood pressure response to the same MC4R agonist was only lost in animals lacking G(s)α specifically in the PVN. Together, our results exemplify how different physiological effects of GPCRs may be mediated by different G proteins and identify a pathway for appetite regulation that could be selectively targeted by G(q/11)α-biased MC4R agonists as a potential treatment for obesity.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Receptor Tipo 4 de Melanocortina/fisiologia , Animais , Colesterol/metabolismo , Feminino , Sistema Hipotálamo-Hipofisário/fisiologia , Resistência à Insulina , Melanocortinas/farmacologia , Camundongos , Camundongos Knockout , Obesidade/etiologia , Sistema Hipófise-Suprarrenal/fisiologia , Receptor Tipo 4 de Melanocortina/agonistas
10.
Artigo em Inglês | MEDLINE | ID: mdl-25261775

RESUMO

Presence of platelet hyperserotonemia and effective amelioration of behavioral dysfunctions by selective serotonin reuptake inhibitors (SSRI) in autism spectrum disorders (ASD) indicate that irregularities in serotonin (5-HT) reuptake and its homeostasis could be the basis of behavioral impairments in ASD patients. SLC6A4, the gene encoding serotonin transporter (SERT) is considered as a potential susceptibility gene for ASD, since it is a quantitative trait locus for blood 5-HT levels. Three functional polymorphisms, 5-HTTLPR, STin2 and 3'UTR-SNP of SLC6A4 are extensively studied for possible association with the disorder, with inconclusive outcome. In the present study, we investigated association of these polymorphisms with platelet 5-HT content and symptoms severity as revealed by childhood autism rating scale in ASD children from an Indian population. Higher 5-HT level observed in ASD was highly significant in children with heterozygous and homozygous genotypes comprising of minor alleles of the markers. Quantitative transmission disequilibrium test demonstrated significant genetic effect of STin2 allele as well as STin2/3'UTR-SNP and 5-HTTLPR/3'UTR-SNP haplotypes on 5-HT levels, but no direct association with overall CARS score and ASD phenotype. Significant genetic effect of the markers on specific behavioral phenotypes was observed for various sub-phenotypes of CARS in quantitative trait analysis. Even though the 5-HT level was not associated with severity of behavioral CARS score, a significant negative relationship was observed for 5-HT levels and level and consistency of intellectual response and general impression in ASD children. Population-based study revealed higher distribution of the haplotype 10/G of STin2/3'UTR-SNP in male controls, suggesting protective effect of this haplotype in male cases. Overall results of the study suggest that SLC6A4 markers have specific genetic effect on individual ASD behavioral attributes, might be through the modulation of 5-HT content.


Assuntos
Transtorno Autístico , Predisposição Genética para Doença/genética , Transtornos Mentais/etiologia , Polimorfismo de Nucleotídeo Único/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Serotonina/sangue , Transtorno Autístico/sangue , Transtorno Autístico/complicações , Transtorno Autístico/genética , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Frequência do Gene , Estudos de Associação Genética , Genótipo , Humanos , Índia/epidemiologia , Masculino , Índice de Gravidade de Doença , Estatística como Assunto , Adulto Jovem
11.
J Biotechnol ; 189: 1-8, 2014 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-25193712

RESUMO

Genomic engineering by the guide RNA (gRNA)-directed CRISPR/CAS9 is rapidly becoming a method of choice for various biological systems. However, pressing concerns remain regarding its off-target activities and wide variations in efficacies. While next generation sequencing (NGS) has been primarily used to evaluate the efficacies and off-target activities of gRNAs, it only detects the imperfectly repaired double strand DNA breaks (DSB) by the error-prone non-homologous end joining (NHEJ) mechanism and may not faithfully represent the DSB activities because the efficiency of NHEJ-mediated repair varies depending on the local chromatin environment. Here we describe a reporter system for unbiased detection and comparison of DSB activities that promises to improve the chance of success in genomic engineering and to facilitate large-scale screening of CAS9 activities and gRNA libraries. Additionally, we demonstrated that the tolerances to mismatches between a gRNA and the corresponding target DNA can occur at any position of the gRNA, and depend on both specific gRNA sequences and CAS9 constructs used.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , DNA/genética , RNA Guia de Cinetoplastídeos/genética , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Humanos
12.
Endocrinology ; 154(1): 550-61, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23161871

RESUMO

Thyroid hormone serves many functions throughout brain development, but the mechanisms that control the timing of its actions in specific brain regions are poorly understood. In the cerebellum, thyroid hormone controls formation of the transient external germinal layer, which contains proliferative granule cell precursors, subsequent granule cell migration, and cerebellar foliation. We report that the thyroid hormone-inactivating type 3 deiodinase (encoded by Dio3) is expressed in the mouse cerebellum at embryonic and neonatal stages, suggesting a need to protect cerebellar tissues from premature stimulation by thyroid hormone. Dio3(-/-) mice displayed reduced foliation, accelerated disappearance of the external germinal layer, and premature expansion of the molecular layer at juvenile ages. Furthermore, Dio3(-/-) mice exhibited locomotor behavioral abnormalities and impaired ability in descending a vertical pole. To ascertain that these phenotypes resulted from inappropriate exposure to thyroid hormone, thyroid hormone receptor α1 (TRα1) was removed from Dio3(-/-) mice, which substantially corrected the cerebellar and behavioral phenotypes. Deletion of TRα1 did not correct the previously reported small thyroid gland or deafness in Dio3(-/-) mice, indicating that Dio3 controls the activation of specific receptor isoforms in different tissues. These findings suggest that type 3 deiodinase constrains the timing of thyroid hormone action during cerebellar development.


Assuntos
Cerebelo/enzimologia , Cerebelo/metabolismo , Iodeto Peroxidase/metabolismo , Receptores alfa dos Hormônios Tireóideos/metabolismo , Animais , Cerebelo/anormalidades , Feminino , Imuno-Histoquímica , Hibridização In Situ , Iodeto Peroxidase/genética , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/genética , Atividade Motora/fisiologia , Receptores alfa dos Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo
14.
J Neurochem ; 119(3): 544-54, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21883221

RESUMO

Gß5 is a divergent member of the signal-transducing G protein ß subunit family encoded by GNB5 and expressed principally in brain and neuronal tissue. Among heterotrimeric Gß isoforms, Gß5 is unique in its ability to heterodimerize with members of the R7 subfamily of the regulator of G protein signaling proteins that contain G protein-γ like domains. Previous studies employing Gnb5 knockout (KO) mice have shown that Gß5 is an essential stabilizer of such regulator of G protein signaling proteins and regulates the deactivation of retinal phototransduction and the proper functioning of retinal bipolar cells. However, little is known of the function of Gß5 in the brain outside the visual system. We show here that mice lacking Gß5 have a markedly abnormal neurologic phenotype that includes impaired development, tiptoe-walking, motor learning and coordination deficiencies, and hyperactivity. We further show that Gß5-deficient mice have abnormalities of neuronal development in cerebellum and hippocampus. We find that the expression of both mRNA and protein from multiple neuronal genes is dysregulated in Gnb5 KO mice. Taken together with previous observations from Gnb5 KO mice, our findings suggest a model in which Gß5 regulates dendritic arborization and/or synapse formation during development, in part by effects on gene expression.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Encéfalo/anormalidades , Encéfalo/crescimento & desenvolvimento , Cerebelo/anormalidades , Subunidades beta da Proteína de Ligação ao GTP/deficiência , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/anormalidades , Anormalidades Múltiplas/fisiopatologia , Animais , Encéfalo/metabolismo , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo
15.
J Bioenerg Biomembr ; 42(3): 217-26, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20464463

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disease which is characterized by psychiatric symptoms, involuntary choreiform movements and dementia with maximum degeneration occurring in striatum and cerebral cortex. Several studies implicate mitochondrial dysfunction to the selective neurodegeneration happening in this disorder. Calcium buffering imbalance and oxidative stress in the mitochondria, critically impaired movement across axons and abnormal fission or fusion of this organelle in the cells are some of the salient features that results in the loss of mitochondrial electron transport chain (ETC) complex function in HD. Although several models involving mutant huntingtin, excitotoxins and mitochondrial complex-II inhibitors have been used to explore the disease, it is not clear how disturbances in mitochondrial functioning is associated with such selective neurodegeneration, or in the expression of huntingtonian phenotypes in animals or man. We have carefully assessed various mitochondrial abnormalities observed in human patient samples, postmortem HD brains, cellular, vertebrate and invertebrate models of the disease, to conclude that ETC dysfunction is an integral part of the disease and justify a causal role of mitochondrial ETC dysfunction for the genesis of this disorder.


Assuntos
Doença de Huntington/patologia , Mitocôndrias/patologia , Animais , Encéfalo/patologia , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Humanos , Doença de Huntington/etiologia , Doença de Huntington/fisiopatologia , Mitocôndrias/fisiologia , Degeneração Neural
16.
Neurochem Int ; 55(6): 431-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19410615

RESUMO

We tested the hypothesis that dopamine contributes significantly to the hydroxyl radical (OH)-induced striatal neurotoxicity caused by 3-nitropropionic acid (3-NP) in a rat model of Huntington's disease. Dopamine (10-100 microM) or 3-NP (10-1000 microM) individually caused a significant increase in the generation of hydroxyl radical (OH) in the mitochondria, which was synergistically enhanced when the lowest dose of the neurotoxin (10 microM) and dopamine (100 microM) were present together. Similarly, systemic administration of l-DOPA (100-250 mg/kg) and a low dose of 3-NP (10 mg/kg) potentiated OH generation in the striatum, and the rats exhibited significant decrease in stride length, a direct indication of neuropathology. The pathology was also evident in striatal sections subjected to NeuN immunohistochemistry. The significant changes in stride length, the production of striatal OH and neuropathological features due to administration of a toxic dose of 3-NP (20 mg/kg) were significantly attenuated by treating the rats with tyrosine hydroxylase inhibitor alpha-methyl-p-tyrosine prior to 3-NP administration. These results strongly implicate a major contributory role of striatal dopamine in increased generation of OH, which leads to striatal neurodegeneration and accompanied behavioral changes, in 3-NP model of Huntington's disease.


Assuntos
Corpo Estriado/metabolismo , Dopamina/metabolismo , Doença de Huntington/metabolismo , Radical Hidroxila/metabolismo , Degeneração Neural/metabolismo , Estresse Oxidativo/fisiologia , Animais , Convulsivantes/toxicidade , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Dopaminérgicos/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Doença de Huntington/induzido quimicamente , Doença de Huntington/fisiopatologia , Coxeadura Animal/induzido quimicamente , Coxeadura Animal/metabolismo , Coxeadura Animal/fisiopatologia , Levodopa/farmacologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Degeneração Neural/induzido quimicamente , Degeneração Neural/fisiopatologia , Neurotoxinas/toxicidade , Nitrocompostos/toxicidade , Fosforilação Oxidativa/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Propionatos/toxicidade , Ratos , Ratos Sprague-Dawley , Tirosina 3-Mono-Oxigenase/antagonistas & inibidores , Tirosina 3-Mono-Oxigenase/metabolismo
17.
J Neurol Sci ; 279(1-2): 39-42, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19176229

RESUMO

The observation of decline in mitochondrial electron transport chain function, specifically at complex I, in patients with Parkinson's disease (PD) has been reported by several groups. This study investigates whether a defect of mitochondrial function is present in the platelets of PD patients from an Indian population. We found that the NADH dehydrogenase activity in the platelets of PD patients is lower than that in healthy age- and gender-matched controls, while the succinate dehydrogenase activity was similar in both groups. Furthermore, there was no change in either of the activities in patients with Parkinson plus syndrome or atypical parkinsonism. This is the first report indicating a decline in mitochondrial function in the platelets of PD patients from the Indian population, offering further support to the role of a mitochondrial defect in PD.


Assuntos
Plaquetas/enzimologia , Complexo I de Transporte de Elétrons/sangue , Doença de Parkinson/enzimologia , Transtornos Parkinsonianos/enzimologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Ativação Enzimática , Feminino , Humanos , Índia , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/sangue , Transtornos Parkinsonianos/sangue , Succinato Desidrogenase/sangue
18.
J Neurochem ; 104(2): 420-34, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17953654

RESUMO

Mitochondrial complex-I dysfunction has been observed in patients of Huntington's disease (HD). We assessed whether such a defect is present in the 3-nitropropionic acid (3-NP) model of HD. Rats treated with 3-NP (10-20 mg/kg i.p., for 4 days) exhibited weight loss, gait abnormalities, and striatal lesions with increased glial fibrillary acidic protein immunostaining on fifth and ninth days, while increase in striatal dopamine and loss of tyrosine hydroxylase immunoreactivity were observed on fifth day following treatment. We report for the first time a dose-dependent reduction in complex-I activity in the cerebral cortex when analyzed spectrophotometrically and by blue native-polyacrylamide gel electrophoresis following 3-NP treatment. The citrate synthase normalized activities of mitochondrial complex-I, -II, -(I + III) and -IV were decreased in the cortex of 3-NP treated rats. In addition, succinate driven State 3 respiration was also significantly inhibited in vivo and in the isolated mitochondria. These findings taken together with the observation of a significant decrease in vivo but not in vitro of State 3 respiration with NAD(+)-linked substrates, suggest complex-I dysfunction in addition to irreversible inhibition of complex-II and succinate dehydrogenase activity as a contributing factor in 3-NP-induced cortico-striatal lesion.


Assuntos
Córtex Cerebral/metabolismo , Doença de Huntington/patologia , Mitocôndrias/enzimologia , Nitrocompostos , Consumo de Oxigênio/fisiologia , Propionatos , Succinato Desidrogenase/metabolismo , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Citrato (si)-Sintase/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Complexo I de Transporte de Elétrons/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Doença de Huntington/induzido quimicamente , Masculino , Mitocôndrias/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
19.
J Clin Microbiol ; 41(11): 5277-81, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14605183

RESUMO

Among Escherichia coli strains isolated from stool specimens from patients with acute diarrhea, 1.4% were found to harbor cdtB by use of enrichment cytolethal distending toxin (CDT) PCR. These isolates were identified as being enteropathogenic E. coli (EPEC). In a retrospective study using a probe hybridization assay, 6 of 138 EPEC strains were found to harbor the cdtB locus. cdtB-positive isolates mostly belong to the O86a and O127a serogroups, with the former being associated with higher expression of CDT. Pulsed-field gel electrophoresis profiles showed that the EPEC strains harboring cdtB strains are genetically diverse.


Assuntos
Toxinas Bacterianas/genética , Diarreia/microbiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Escherichia coli/patogenicidade , Fezes/microbiologia , Doença Aguda , Toxinas Bacterianas/classificação , Toxinas Bacterianas/isolamento & purificação , Sequência de Bases , Sangue/microbiologia , Primers do DNA , Desidratação/microbiologia , Eletroforese em Gel de Campo Pulsado , Escherichia coli/classificação , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/sangue , Proteínas de Escherichia coli/isolamento & purificação , Humanos , Índia , Dados de Sequência Molecular , Sangue Oculto , Filogenia , Reação em Cadeia da Polimerase , Sorotipagem/métodos , Vômito/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA