Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 7(12): e08518, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34926857

RESUMO

Neurological diseases, such as MS, AD, PD and HD, are a major health concern of the elderly population, but still therapeutic options are limited. Recent advances in genomic sequencing and bioinformatics, present an opportunity to understand mechanisms of these diseases for identification of therapeutic targets. Several studies have shown association of immune dysfunction with immune system mediated neurological disease, MS, as well as neurodegenerative diseases (AD, PD and HD). However, similarities and differences in role of the immune system, immune pathways and immune cell types in these diseases remains unknown. In this study, immune cell type signature genes in gene networks associated with neurological diseases, MS, AD, PD and HD was investigated using meta-analysis and bioinformatics methods. Application of Weighted Gene Co-expression Network Analysis (WGCNA) on publicly available gene expression datasets (microarray and RNA-seq) revealed a ModArray_04 module (microarray) or ModRNAseq_06 module (RNA-seq), significantly associated with MS, AD, PD and HD. Hypergeometric enrichment test revealed significant enrichment of immune cell type genes in these neurological disease modules. This study demonstrates that immune system mediated neurological disease, MS and neurodegenerative diseases (AD, PD and HD), share a common gene network characterized by immune cell type signature genes (microglia, monocytes and macrophages) and are probable targets for therapeutic intervention. In summary, this work shows a connection between MS, a disease where the role of the immune system and inflammation is established, and neurodegenerative diseases (AD, PD and HD) where the role of inflammation is still a hypothesis.

2.
Sci Rep ; 10(1): 10937, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616845

RESUMO

Grade 4 glioma or GBM has poor prognosis and is the most aggressive grade of glioma. Accurate diagnosis and classification of tumor grade is a critical determinant for development of treatment pathway. Extensive genomic sequencing of gliomas, different cell types, brain tissue regions and advances in bioinformatics algorithms, have presented an opportunity to identify molecular markers that can complement existing histology and imaging methods used to diagnose and classify gliomas. 'Cancer stem cell theory' purports that a minor population of stem cells among the heterogeneous population of different cell types in the tumor, drive tumor growth and resistance to therapies. However, characterization of stem cell states in GBM and ability of stem cell state signature genes to serve as diagnostic or prognostic molecular markers are unknown. In this work, two different network construction algorithms, Weighted correlation network analysis (WGCNA) and Multiscale Clustering of Geometric Network (MEGENA), were applied on publicly available glioma, control brain and stem cell gene expression RNA-seq datasets, to identify gene network regulatory modules associated with GBM. Both gene network algorithms identified consensus or equivalent modules, HuAgeGBsplit_18 (WGCNA) and c1_HuAgeGBsplit_32/193 (MEGENA), significantly associated with GBM. Characterization of HuAgeGBsplit_18 (WGCNA) and c1_HuAgeGBsplit_32/193 (MEGENA) modules showed significant enrichment of rodent quiescent stem cell marker genes (GSE70696_QNPbyTAP). A logistic regression model built with eight of these quiescent stem cell marker genes (GSE70696_QNPbyTAP) was sufficient to distinguish between control and GBM samples. This study demonstrates that GBM associated gene regulatory modules are characterized by diagnostic quiescent stem cell marker genes, which may potentially be used clinically as diagnostic markers and therapeutic targets in GBM.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Encéfalo/patologia , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Estudos de Casos e Controles , Biologia Computacional , Glioblastoma/genética , Humanos , Células-Tronco Neoplásicas/metabolismo , Prognóstico
3.
Cancer Biother Radiopharm ; 34(8): 487-497, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31314580

RESUMO

Next-generation sequencing (NGS) data have been central to the development of targeted therapy and immunotherapy for precision oncology. In targeted therapy, drugs directly attack cancer, by altering the expression of critical cancer genes identified with cancer genome profiling. Immunotherapy drugs indirectly attack cancer, by inducing the immune system to attack and treat cancer. Harnessing genomic data for deployment and development of immunotherapy comprises the field of immunogenomics. The discovery of a link between cancer cells escaping immune destruction and cancer progression, led to extensive research into this mechanism and drug development. In the past few years, FDA has granted accelerated approval to several immunotherapy cancer treatment drugs, pembrolizumab, nivolumab, and atezolizumab, belonging to the class of checkpoint inhibitors. Utilization of pretreatment genomic cancer screening to identify patients most likely to respond to immunotherapy and to customize immunotherapy for a given patient, promises to improve cancer treatment outcomes. Recent advances in molecular profiling, high-throughput sequencing, and computational efficiency has made immunogenomics the major tenet of precision medicine in cancer treatment. This review provides a brief overview on the state of art of immunogenomics in precision cancer medicine.


Assuntos
Genômica/métodos , Imunoterapia/métodos , Neoplasias/terapia , Medicina de Precisão/métodos , Biomarcadores Tumorais/genética , Humanos , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/imunologia
4.
Front Neurosci ; 13: 2, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30733664

RESUMO

Aging is regarded as a major risk factor for neurodegenerative diseases. Thus, a better understanding of the similarities between the aging process and neurodegenerative diseases at the cellular and molecular level may reveal better understanding of this detrimental relationship. In the present study, we mined publicly available gene expression datasets from healthy individuals and patients affected by neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, and Huntington's disease) across a broad age spectrum and compared those with mouse aging and mouse cell-type specific gene expression profiles. We performed weighted gene co-expression network analysis (WGCNA) and found a gene network strongly related with both aging and neurodegenerative diseases. This network was significantly enriched with a microglial signature as imputed from cell type-specific sequencing data. Since mouse models are extensively used for the study of human diseases, we further compared these human gene regulatory networks with age-specific mouse brain transcriptomes. We discovered significantly preserved networks with both human aging and human disease and identified 17 shared genes in the top-ranked immune/microglia module, among which we found five human hub genes TYROBP, FCER1G, ITGB2, MYO1F, PTPRC, and two mouse hub genes Trem2 and C1qa. Taken together, these results support the hypothesis that microglia are key players involved in human aging and neurodegenerative diseases, and suggest that mouse models should be appropriate for studying these microglial changes in human.

5.
Methods Mol Biol ; 1686: 265-286, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29030827

RESUMO

Transcription factors bind to specific DNA sequences and control the transcription rate of nearby genes in the genome. This activation or repression of gene expression is further potentiated by epigenetic modifications of histones with active and silent marks, respectively. Resident adult stem cells in the hematopoietic system, skin, and brain exist in a non-proliferative quiescent resting state. When quiescent stem cells become activated and transition to dividing progenitors and distinct cell types, they can replenish and repair tissue. Thus, determination of the position of transcription factor binding and histone epigenetic modification on the chromatin is an essential step toward understanding the gene regulation of quiescent and proliferative adult stem cells for potential applications in regenerative medicine. Genome-wide transcription factor occupancy and histone modifications on the genome can be obtained by assessing DNA-protein interaction through next-generation chromatin immunoprecipitation sequencing technology (ChIP-seq). This chapter outlines the protocol to perform, analyze, and validate ChIP-seq experiments that can be used to identify protein-DNA interactions and histone marks on the chromatin. The methods described here are applicable to quiescent and proliferative neural stem cells, and a wide range of other cellular systems.


Assuntos
Células-Tronco Adultas/metabolismo , Imunoprecipitação da Cromatina/métodos , Cromatina/metabolismo , Genoma Humano , Células-Tronco Neurais/metabolismo , Fase de Repouso do Ciclo Celular , Fatores de Transcrição/metabolismo , Adulto , Células-Tronco Adultas/citologia , Sítios de Ligação , Células Cultivadas , Cromatina/genética , Epigênese Genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Histonas/genética , Histonas/metabolismo , Humanos , Células-Tronco Neurais/citologia , Ligação Proteica , Análise de Sequência de DNA/métodos , Fatores de Transcrição/genética
6.
Nat Commun ; 7: 13360, 2016 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-27819263

RESUMO

Adult hippocampal neural stem cells generate newborn neurons throughout life due to their ability to self-renew and exist as quiescent neural progenitors (QNPs) before differentiating into transit-amplifying progenitors (TAPs) and newborn neurons. The mechanisms that control adult neural stem cell self-renewal are still largely unknown. Conditional knockout of REST (repressor element 1-silencing transcription factor) results in precocious activation of QNPs and reduced neurogenesis over time. To gain insight into the molecular mechanisms by which REST regulates adult neural stem cells, we perform chromatin immunoprecipitation sequencing and RNA-sequencing to identify direct REST target genes. We find REST regulates both QNPs and TAPs, and importantly, ribosome biogenesis, cell cycle and neuronal genes in the process. Furthermore, overexpression of individual REST target ribosome biogenesis or cell cycle genes is sufficient to induce activation of QNPs. Our data define novel REST targets to maintain the quiescent neural stem cell state.


Assuntos
Células-Tronco Adultas/fisiologia , Redes Reguladoras de Genes/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Proteínas Repressoras/fisiologia , Animais , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Imunoprecipitação da Cromatina/métodos , Técnicas de Inativação de Genes , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Hipocampo/citologia , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Neurônios/fisiologia , Proteínas Repressoras/genética , Ribossomos/fisiologia , Análise de Sequência de RNA
7.
Mol Vis ; 22: 1468-1489, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28050121

RESUMO

PURPOSE: Studies of neuronal development in the retina often examine the stages of proliferation, differentiation, and synaptic development, albeit independently. Our goal was to determine if a known neurotoxicant insult to a population of retinal progenitor cells (RPCs) would affect their eventual differentiation and synaptic development. To that end, we used our previously published human equivalent murine model of low-level gestational lead exposure (GLE). Children and animals with GLE exhibit increased scotopic electroretinogram a- and b-waves. Adult mice with GLE exhibit an increased number of late-born RPCs, a prolonged period of RPC proliferation, and an increased number of late-born rod photoreceptors and rod and cone bipolar cells (BCs), with no change in the number of late-born Müller glial cells or early-born neurons. The specific aims of this study were to determine whether increased and prolonged RPC proliferation alters the spatiotemporal differentiation and synaptic development of rods and BCs in early postnatal GLE retinas compared to control retinas. METHODS: C57BL/6N mouse pups were exposed to lead acetate via drinking water throughout gestation and until postnatal day 10, which is equivalent to the human gestation period for retinal neurogenesis. RT-qPCR, immunohistochemical analysis, and western blots of well-characterized, cell-specific genes and proteins were performed at embryonic and early postnatal ages to assess rod and cone photoreceptor differentiation, rod and BC differentiation and synaptic development, and Müller glial cell differentiation. RESULTS: Real-time quantitative PCR (RT-qPCR) with the rod-specific transcription factors Nrl, Nr2e3, and Crx and the rod-specific functional gene Rho, along with central retinal confocal studies with anti-recoverin and anti-rhodopsin antibodies, revealed a two-day delay in the differentiation of rod photoreceptors in GLE retinas. Rhodopsin immunoblots supported this conclusion. No changes in glutamine synthetase gene or protein expression, a marker for late-born Müller glial cells, were observed in the developing retinas. In the retinas from the GLE mice, anti-PKCα, -Chx10 (Vsx2) and -secretagogin antibodies revealed a two- to three-day delay in the differentiation of rod and cone BCs, whereas the expression of the proneural and BC genes Otx2 and Chx10, respectively, increased. In addition, confocal studies of proteins associated with functional synapses (e.g., vesicular glutamate transporter 1 [VGluT1], plasma membrane calcium ATPase [PMCA], transient receptor potential channel M1 [TRPM1], and synaptic vesicle glycoprotein 2B [SV2B]) revealed a two-day delay in the formation of the outer and inner plexiform layers of the GLE retinas. Moreover, several markers revealed that the initiation of the differentiation and intensity of the labeling of early-born cells in the retinal ganglion cell and inner plexiform layers were not different in the control retinas. CONCLUSIONS: Our combined gene, confocal, and immunoblot findings revealed that the onset of rod and BC differentiation and their subsequent synaptic development is delayed by two to three days in GLE retinas. These results suggest that perturbations during the early proliferative stages of late-born RPCs fated to be rods and BCs ultimately alter the coordinated time-dependent progression of rod and BC differentiation and synaptic development. These GLE effects were selective for late-born neurons. Although the molecular mechanisms are unknown, alterations in soluble neurotrophic factors and/or their receptors are likely to play a role. Since neurodevelopmental delays and altered synaptic connectivity are associated with neuropsychiatric and behavioral disorders as well as cognitive deficits, future work is needed to determine if similar effects occur in the brains of GLE mice and whether children with GLE experience similar delays in retinal and brain neuronal differentiation and synaptic development.


Assuntos
Diferenciação Celular , Chumbo/toxicidade , Neurogênese , Efeitos Tardios da Exposição Pré-Natal/patologia , Células Bipolares da Retina/patologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Envelhecimento/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glutamato-Amônia Ligase/metabolismo , Camundongos Endogâmicos C57BL , Neurogênese/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos Sprague-Dawley , Células Bipolares da Retina/efeitos dos fármacos , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Rodopsina/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
8.
Cell Stem Cell ; 13(2): 133-4, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23910076

RESUMO

Tet proteins and DNA demethylation are key regulators of embryonic stem cells, but their roles in development and tissue stem cells remain unclear. In this issue, Zhang et al. (2013) show that Tet1 regulates adult hippocampal neurogenesis by controlling progenitor proliferation, and Tet1 deficiency generates defects in learning and memory.


Assuntos
Envelhecimento/metabolismo , Cognição , Proteínas de Ligação a DNA/metabolismo , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Neurogênese , Proteínas Proto-Oncogênicas/metabolismo , Animais
9.
Toxicol Appl Pharmacol ; 256(3): 258-67, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21703292

RESUMO

Gestational lead exposure (GLE) produces supernormal scotopic electroretinograms (ERG) in children, monkeys and rats, and a novel retinal phenotype characterized by an increased number of rod photoreceptors and bipolar cells in adult mice and rats. Since the loss of dopaminergic amacrine cells (DA ACs) in GLE monkeys and rats contributes to supernormal ERGs, the retinal DA system was analyzed in mice following GLE. C57BL/6 female mice were exposed to low (27 ppm), moderate (55 ppm) or high (109 ppm) lead throughout gestation and until postnatal day 10 (PN10). Blood [Pb] in control, low-, moderate- and high-dose GLE was ≤ 1, ≤ 10, ~25 and ~40 µg/dL, respectively, on PN10 and by PN30 all were ≤ 1 µg/dL. At PN60, confocal-stereology studies used vertical sections and wholemounts to characterize tyrosine hydroxylase (TH) expression and the number of DA and other ACs. GLE dose-dependently and selectively decreased the number of TH-immunoreactive (IR) DA ACs and their synaptic plexus without affecting GABAergic, glycinergic or cholinergic ACs. Immunoblots and confocal revealed dose-dependent decreases in retinal TH protein expression and content, although monoamine oxidase-A protein and gene expression were unchanged. High-pressure liquid chromatography showed that GLE dose-dependently decreased retinal DA content, its metabolites and DA utilization/release. The mechanism of DA selective vulnerability is unknown. However, a GLE-induced loss/dysfunction of DA ACs during development could increase the number of rods and bipolar cells since DA helps regulate neuronal proliferation, whereas during adulthood it could produce ERG supernormality as well as altered circadian rhythms, dark/light adaptation and spatial contrast sensitivity.


Assuntos
Células Amácrinas/efeitos dos fármacos , Dopamina/análise , Intoxicação do Sistema Nervoso por Chumbo/metabolismo , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Células Amácrinas/química , Células Amácrinas/patologia , Animais , Western Blotting , Contagem de Células , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Feminino , Intoxicação do Sistema Nervoso por Chumbo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Reação em Cadeia da Polimerase em Tempo Real
10.
Environ Health Perspect ; 119(1): 71-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20840909

RESUMO

BACKGROUND: Gestational lead exposure (GLE) produces novel and persistent rod-mediated electroretinographic (ERG) supernormality in children and adult animals. OBJECTIVES: We used our murine GLE model to test the hypothesis that GLE increases the number of neurons in the rod signaling pathway and to determine the cellular mechanisms underlying the phenotype. RESULTS: Blood lead concentrations ([BPb]) in controls and after low-, moderate-, and high-dose GLE were ≤ 1, ≤ 10, approximately 25, and approximately 40 µg/dL, respectively, at the end of exposure [postnatal day 10 (PND10)]; by PND30 all [BPb] measures were ≤ 1 µg/dL. Epifluorescent, light, and confocal microscopy studies and Western blots demonstrated that late-born rod photoreceptors and rod and cone bipolar cells (BCs), but not Müller glial cells, increased in a nonmonotonic manner by 16-30% in PND60 GLE offspring. Retinal lamination and the rod:cone BC ratio were not altered. In vivo BrdU (5-bromo-2-deoxyuridine) pulse-labeling and Ki67 labeling of isolated cells from developing mice showed that GLE increased and prolonged retinal progenitor cell proliferation. TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) and confocal studies revealed that GLE did not alter developmental apoptosis or produce retinal injury. BrdU birth-dating and confocal studies confirmed the selective rod and BC increases and showed that the patterns of neurogenesis and gliogenesis were unaltered by GLE. CONCLUSIONS: Our findings suggest two spatiotemporal components mediated by dysregulation of different extrinsic/intrinsic factors: increased and prolonged cell proliferation and increased neuronal (but not glial) cell fate. These findings have relevance for neurotoxicology, pediatrics, public health, risk assessment, and retinal cell biology because they occurred at clinically relevant [BPb] and correspond with the ERG phenotype.


Assuntos
Poluentes Ambientais/toxicidade , Chumbo/toxicidade , Retina/efeitos dos fármacos , Células Bipolares da Retina/efeitos dos fármacos , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Poluentes Ambientais/sangue , Feminino , Chumbo/sangue , Masculino , Exposição Materna , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neurogênese/efeitos dos fármacos , Retina/crescimento & desenvolvimento , Células Bipolares da Retina/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...