Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
PLoS Genet ; 18(7): e1009977, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35788729

RESUMO

African descent populations have a lower Alzheimer disease risk from ApoE ε4 compared to other populations. Ancestry analysis showed that the difference in risk between African and European populations lies in the ancestral genomic background surrounding the ApoE locus (local ancestry). Identifying the mechanism(s) of this protection could lead to greater insight into the etiology of Alzheimer disease and more personalized therapeutic intervention. Our objective is to follow up the local ancestry finding and identify the genetic variants that drive this risk difference and result in a lower risk for developing Alzheimer disease in African ancestry populations. We performed association analyses using a logistic regression model with the ApoE ε4 allele as an interaction term and adjusted for genome-wide ancestry, age, and sex. Discovery analysis included imputed SNP data of 1,850 Alzheimer disease and 4,331 cognitively intact African American individuals. We performed replication analyses on 63 whole genome sequenced Alzheimer disease and 648 cognitively intact Ibadan individuals. Additionally, we reproduced results using whole-genome sequencing of 273 Alzheimer disease and 275 cognitively intact admixed Puerto Rican individuals. A further comparison was done with SNP imputation from an additional 8,463 Alzheimer disease and 11,365 cognitively intact non-Hispanic White individuals. We identified a significant interaction between the ApoE ε4 allele and the SNP rs10423769_A allele, (ß = -0.54,SE = 0.12,p-value = 7.50x10-6) in the discovery data set, and replicated this finding in Ibadan (ß = -1.32,SE = 0.52,p-value = 1.15x10-2) and Puerto Rican (ß = -1.27,SE = 0.64,p-value = 4.91x10-2) individuals. The non-Hispanic Whites analyses showed an interaction trending in the "protective" direction but failing to pass a 0.05 significance threshold (ß = -1.51,SE = 0.84,p-value = 7.26x10-2). The presence of the rs10423769_A allele reduces the odds ratio for Alzheimer disease risk from 7.2 for ApoE ε4/ε4 carriers lacking the A allele to 2.1 for ApoE ε4/ε4 carriers with at least one A allele. This locus is located approximately 2 mB upstream of the ApoE locus, in a large cluster of pregnancy specific beta-1 glycoproteins on chromosome 19 and lies within a long noncoding RNA, ENSG00000282943. This study identified a new African-ancestry specific locus that reduces the risk effect of ApoE ε4 for developing Alzheimer disease. The mechanism of the interaction with ApoEε4 is not known but suggests a novel mechanism for reducing the risk for ε4 carriers opening the possibility for potential ancestry-specific therapeutic intervention.


Assuntos
Doença de Alzheimer , Alelos , Doença de Alzheimer/genética , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Genótipo , Humanos , Nigéria , Fatores de Risco
2.
Hum Genet ; 143(5): 721-734, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38691166

RESUMO

TMPRSS3-related hearing loss presents challenges in correlating genotypic variants with clinical phenotypes due to the small sample sizes of previous studies. We conducted a cross-sectional genomics study coupled with retrospective clinical phenotype analysis on 127 individuals. These individuals were from 16 academic medical centers across 6 countries. Key findings revealed 47 unique TMPRSS3 variants with significant differences in hearing thresholds between those with missense variants versus those with loss-of-function genotypes. The hearing loss progression rate for the DFNB8 subtype was 0.3 dB/year. Post-cochlear implantation, an average word recognition score of 76% was observed. Of the 51 individuals with two missense variants, 10 had DFNB10 with profound hearing loss. These 10 all had at least one of 4 TMPRSS3 variants predicted by computational modeling to be damaging to TMPRSS3 structure and function. To our knowledge, this is the largest study of TMPRSS3 genotype-phenotype correlations. We find significant differences in hearing thresholds, hearing loss progression, and age of presentation, by TMPRSS3 genotype and protein domain affected. Most individuals with TMPRSS3 variants perform well on speech recognition tests after cochlear implant, however increased age at implant is associated with worse outcomes. These findings provide insight for genetic counseling and the on-going design of novel therapeutic approaches.


Assuntos
Estudos de Associação Genética , Perda Auditiva , Proteínas de Membrana , Serina Endopeptidases , Humanos , Feminino , Masculino , Serina Endopeptidases/genética , Adulto , Proteínas de Membrana/genética , Perda Auditiva/genética , Criança , Pessoa de Meia-Idade , Adolescente , Pré-Escolar , Genótipo , Estudos de Coortes , Fenótipo , Mutação de Sentido Incorreto , Estudos Transversais , Adulto Jovem , Estudos Retrospectivos , Idoso , Proteínas de Neoplasias
3.
Cell ; 133(4): 681-92, 2008 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-18485875

RESUMO

The killer lymphocyte protease granzyme A (GzmA) triggers caspase-independent target cell death with morphological features of apoptosis. We previously showed that GzmA acts directly on mitochondria to generate reactive oxygen species (ROS) and disrupt the transmembrane potential (DeltaPsi(m)) but does not permeabilize the mitochondrial outer membrane. Mitochondrial damage is critical to GzmA-induced cell death since cells treated with superoxide scavengers are resistant to GzmA. Here we find that GzmA accesses the mitochondrial matrix to cleave the complex I protein NDUFS3, an iron-sulfur subunit of the NADH:ubiquinone oxidoreductase complex I, after Lys56 to interfere with NADH oxidation and generate superoxide anions. Target cells expressing a cleavage site mutant of NDUFS3 are resistant to GzmA-mediated cell death but remain sensitive to GzmB.


Assuntos
Morte Celular , Granzimas/metabolismo , Mitocôndrias/química , Mitocôndrias/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Sistema Livre de Células , Granzimas/genética , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Humanos , Fígado/citologia , Potencial da Membrana Mitocondrial , Camundongos , NADH Desidrogenase/metabolismo , Oxirredutases/metabolismo , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/metabolismo , Rotenona/farmacologia , Linfócitos T Citotóxicos/enzimologia , Desacopladores/farmacologia
4.
Alzheimers Dement ; 19(9): 3902-3915, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37037656

RESUMO

INTRODUCTION: European local ancestry (ELA) surrounding apolipoprotein E (APOE) ε4 confers higher risk for Alzheimer's disease (AD) compared to African local ancestry (ALA). We demonstrated significantly higher APOE ε4  expression in ELA versus ALA in AD brains from APOE ε4/ε4 carriers. Chromatin accessibility differences could contribute to these expression changes. METHODS: We performed single nuclei assays for transposase accessible chromatin sequencing from the frontal cortex of six ALA and six ELA AD brains, homozygous for local ancestry and APOE ε4. RESULTS: Our results showed an increased chromatin accessibility at the APOE ε4  promoter area in ELA versus ALA astrocytes. This increased accessibility in ELA astrocytes extended genome wide. Genes with increased accessibility in ELA in astrocytes were enriched for synapsis, cholesterol processing, and astrocyte reactivity. DISCUSSION: Our results suggest that increased chromatin accessibility of APOE ε4  in ELA astrocytes contributes to the observed elevated APOE ε4  expression, corresponding to the increased AD risk in ELA versus ALA APOE ε4/ε4 carriers.


Assuntos
Doença de Alzheimer , Apolipoproteína E4 , Humanos , Apolipoproteína E4/genética , Doença de Alzheimer/genética , Doença de Alzheimer/complicações , Cromatina , Heterozigoto , Expressão Gênica
5.
Int J Mol Sci ; 24(7)2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37047559

RESUMO

Cocaine abuse increases the risk of atherosclerotic cardiovascular disease (CVD) and causes acute coronary syndromes (ACS) and hypertension (HTN). Significant research has explored the role of the sympathetic nervous system mediating the cocaine effects on the cardiovascular (CV) system. However, the response of the sympathetic nervous system alone is insufficient to completely account for the CV consequences seen in cocaine users. In this study, we examined the role of microRNAs (miRNAs) in mediating the effect of cocaine on the CV system. MiRNAs regulate many important biological processes and have been associated with both response to cocaine and CV disease development. Multiple miRNAs have altered expression in the CV system (CVS) upon cocaine exposure. To understand the molecular mechanisms underlying the cocaine response in the CV system, we studied the role of miRNA-423-5p and its target Cacna2d2 in the regulation of intracellular calcium concentration and SMC contractility, a critical factor in the modulation of blood pressure (BP). We used in vivo models to evaluate BP and aortic stiffness. In vitro, cocaine treatment decreased miR-423-5p expression and increased Cacna2d2 expression, which led to elevated intracellular calcium concentrations and increased SMC contractility. Overexpression of miR-423-5p, silencing of its target Cacna2d2, and treatment with a calcium channel blocker reversed the elevated SMC contractility caused by cocaine. In contrast, suppression of miR-423-5p increased the intracellular calcium concentration and SMC contractibility. In vivo, smooth muscle-specific overexpression of miR-423-5p ameliorated the increase in BP and aortic stiffness associated with cocaine use. Thus, miR-423-5p regulates SMC contraction by modulating Cacna2d2 expression increasing intracellular calcium concentrations. Modulation of the miR-423-5p-Cacna2d2-Calcium transport pathway may represent a novel therapeutic strategy to improve cocaine-induced HTN and aortic stiffness.


Assuntos
Aterosclerose , Transtornos Relacionados ao Uso de Cocaína , Cocaína , MicroRNAs , Humanos , Cocaína/efeitos adversos , Cocaína/metabolismo , Cálcio/metabolismo , MicroRNAs/metabolismo , Aterosclerose/metabolismo , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Miócitos de Músculo Liso/metabolismo , Proliferação de Células , Canais de Cálcio/metabolismo
6.
Int J Mol Sci ; 24(8)2023 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-37108729

RESUMO

People living with HIV (PLHIV) are at a higher risk of having cerebrocardiovascular diseases (CVD) compared to HIV negative (HIVneg) individuals. The mechanisms underlying this elevated risk remains elusive. We hypothesize that HIV infection results in modified microRNA (miR) content in plasma extracellular vesicles (EVs), which modulates the functionality of vascular repairing cells, i.e., endothelial colony-forming cells (ECFCs) in humans or lineage negative bone marrow cells (lin- BMCs) in mice, and vascular wall cells. PLHIV (N = 74) have increased atherosclerosis and fewer ECFCs than HIVneg individuals (N = 23). Plasma from PLHIV was fractionated into EVs (HIVposEVs) and plasma depleted of EVs (HIV PLdepEVs). HIVposEVs, but not HIV PLdepEVs or HIVnegEVs (EVs from HIVneg individuals), increased atherosclerosis in apoE-/- mice, which was accompanied by elevated senescence and impaired functionality of arterial cells and lin- BMCs. Small RNA-seq identified EV-miRs overrepresented in HIVposEVs, including let-7b-5p. MSC (mesenchymal stromal cell)-derived tailored EVs (TEVs) loaded with the antagomir for let-7b-5p (miRZip-let-7b) counteracted, while TEVs loaded with let-7b-5p recapitulated the effects of HIVposEVs in vivo. Lin- BMCs overexpressing Hmga2 (a let-7b-5p target gene) lacking the 3'UTR and as such is resistant to miR-mediated regulation showed protection against HIVposEVs-induced changes in lin- BMCs in vitro. Our data provide a mechanism to explain, at least in part, the increased CVD risk seen in PLHIV.


Assuntos
Aterosclerose , MicroRNA Circulante , Vesículas Extracelulares , Infecções por HIV , MicroRNAs , Humanos , Animais , Camundongos , Infecções por HIV/complicações , Infecções por HIV/genética , MicroRNAs/genética , Vesículas Extracelulares/genética , Aterosclerose/genética
7.
Alzheimers Dement ; 18(10): 1930-1942, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34978147

RESUMO

We previously demonstrated that in Alzheimer's disease (AD) patients, European apolipoprotein E (APOE) ε4 carriers express significantly more APOE ε4 in their brains than African AD carriers. We examined single nucleotide polymorphisms near APOE with significant frequency differences between African and European/Japanese APOE ε4 haplotypes that could contribute to this difference in expression through regulation. Two enhancer massively parallel reporter assay (MPRA) approaches were performed, supplemented with single fragment reporter assays. We used Capture C analyses to support interactions with the APOE promoter. Introns within TOMM40 showed increased enhancer activity in the European/Japanese versus African haplotypes in astrocytes and microglia. This region overlaps with APOE promoter interactions as assessed by Capture C analysis. Single variant analyses pinpoints rs2075650/rs157581, and rs59007384 as functionally different on these haplotypes. Identification of the mechanisms for differential regulatory function for APOE expression between African and European/Japanese haplotypes could lead to therapeutic targets for APOE ε4 carriers.


Assuntos
Doença de Alzheimer , Apolipoproteína E4 , Humanos , Alelos , Doença de Alzheimer/genética , Apolipoproteína E4/genética , Apolipoproteínas E/genética , População Negra/genética , Genótipo , Haplótipos , Polimorfismo de Nucleotídeo Único/genética
8.
PLoS Pathog ; 15(7): e1007883, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31260499

RESUMO

Chronic infection with human immunodeficiency virus (HIV) and hepatitis C virus (HCV) affects an estimated 35 million and 75 million individuals worldwide, respectively. These viruses induce persistent inflammation which often drives the development or progression of organ-specific diseases and even cancer including Hepatocellular Carcinoma (HCC). In this study, we sought to examine inflammatory responses following HIV or HCV stimulation of macrophages or Kupffer cells (KCs), that may contribute to virus mediated inflammation and subsequent liver disease. KCs are liver-resident macrophages and reports have provided evidence that HIV can stimulate and infect them. In order to characterize HIV-intrinsic innate immune responses that may occur in the liver, we performed microarray analyses on KCs following HIV stimulation. Our data demonstrate that KCs upregulate several innate immune signaling pathways involved in inflammation, myeloid cell maturation, stellate cell activation, and Triggering Receptor Expressed on Myeloid cells 1 (TREM1) signaling. TREM1 is a member of the immunoglobulin superfamily of receptors and it is reported to be involved in systemic inflammatory responses due to its ability to amplify activation of host defense signaling pathways. Our data demonstrate that stimulation of KCs with HIV or HCV induces the upregulation of TREM1. Additionally, HIV viral proteins can upregulate expression of TREM1 mRNA through NF-кB signaling. Furthermore, activation of the TREM1 signaling pathway, with a targeted agonist, increased HIV or HCV-mediated inflammatory responses in macrophages due to enhanced activation of the ERK1/2 signaling cascade. Silencing TREM1 dampened inflammatory immune responses elicited by HIV or HCV stimulation. Finally, HIV and HCV infected patients exhibit higher expression and frequency of TREM1 and CD68 positive cells. Taken together, TREM1 induction by HIV contributes to chronic inflammation in the liver and targeting TREM1 signaling may be a therapeutic option to minimize HIV induced chronic inflammation.


Assuntos
Infecções por HIV/imunologia , Hepatite C Crônica/imunologia , Receptor Gatilho 1 Expresso em Células Mieloides/imunologia , Estudos de Casos e Controles , Linhagem Celular , Quimiocinas/biossíntese , Citocinas/biossíntese , Infecções por HIV/complicações , Infecções por HIV/genética , Hepatite C Crônica/complicações , Hepatite C Crônica/genética , Humanos , Imunidade Inata/genética , Inflamação/etiologia , Inflamação/genética , Inflamação/imunologia , Células de Kupffer/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Células Mieloides/imunologia , Transdução de Sinais/imunologia , Receptor Gatilho 1 Expresso em Células Mieloides/genética
9.
PLoS Genet ; 14(10): e1007667, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30289878

RESUMO

The role of host genetic variation in the development of complicated Staphylococcus aureus bacteremia (SAB) is poorly understood. We used whole exome sequencing (WES) to examine the cumulative effect of coding variants in each gene on risk of complicated SAB in a discovery sample of 168 SAB cases (84 complicated and 84 uncomplicated, frequency matched by age, sex, and bacterial clonal complex [CC]), and then evaluated the most significantly associated genes in a replication sample of 240 SAB cases (122 complicated and 118 uncomplicated, frequency matched for age, sex, and CC) using targeted sequence capture. In the discovery sample, gene-based analysis using the SKAT-O program identified 334 genes associated with complicated SAB at p<3.5 x 10-3. These, along with eight biologically relevant candidate genes were examined in the replication sample. Gene-based analysis of the 342 genes in the replication sample using SKAT-O identified one gene, GLS2, significantly associated with complicated SAB (p = 1.2 x 10-4) after Bonferroni correction. In Firth-bias corrected logistic regression analysis of individual variants, the strongest association across all 10,931 variants in the replication sample was with rs2657878 in GLS2 (p = 5 x 10-4). This variant is strongly correlated with a missense variant (rs2657879, p = 4.4 x 10-3) in which the minor allele (associated here with complicated SAB) has been previously associated with lower plasma concentration of glutamine. In a microarray-based gene-expression analysis, individuals with SAB exhibited significantly lower expression levels of GLS2 than healthy controls. Similarly, Gls2 expression is lower in response to S. aureus exposure in mouse RAW 264.7 macrophage cells. Compared to wild-type cells, RAW 264.7 cells with Gls2 silenced by CRISPR-Cas9 genome editing have decreased IL1-ß transcription and increased nitric oxide production after S. aureus exposure. GLS2 is an interesting candidate gene for complicated SAB due to its role in regulating glutamine metabolism, a key factor in leukocyte activation.


Assuntos
Glutaminase/genética , Infecções Estafilocócicas/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Animais , Bacteriemia , Feminino , Frequência do Gene/genética , Variação Genética/genética , Glutaminase/metabolismo , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Células RAW 264.7 , Fatores de Risco , Staphylococcus aureus/patogenicidade , Transcriptoma/genética , Sequenciamento do Exoma/métodos
10.
Alzheimers Dement ; 17(7): 1179-1188, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33522086

RESUMO

INTRODUCTION: Apolipoprotein E (APOE) ε4 confers less risk for Alzheimer's disease (AD) in carriers with African local genomic ancestry (ALA) than APOE ε4 carriers with European local ancestry (ELA). Cell type specific transcriptional variation between the two local ancestries (LAs) could contribute to this disease risk differences. METHODS: Single-nucleus RNA sequencing was performed on frozen frontal cortex of homozygous APOE ε4/ε4 AD patients: seven with ELA, four with ALA. RESULTS: A total of 60,908 nuclei were sequenced. Within the LA region (chr19:44-46Mb), APOE was the gene most differentially expressed, with ELA carriers having significantly more expression (overall P < 1.8E-317 ) in 24 of 32 cell clusters. The transcriptome of one astrocyte cluster, with high APOE ε4 expression and specific to ELA, is suggestive of A1 reactive astrocytes. DISCUSSION: AD patients with ELA expressed significantly greater levels of APOE than ALA APOE ε4 carriers. These differences in APOE expression could contribute to the reduced risk for AD seen in African APOE ε4 carriers.


Assuntos
Doença de Alzheimer , Apolipoproteína E4/genética , População Negra/genética , Análise de Sequência de RNA , População Branca/genética , Idoso , Idoso de 80 Anos ou mais , Alelos , Doença de Alzheimer/etnologia , Doença de Alzheimer/genética , Feminino , Heterozigoto , Humanos , Masculino
11.
Int J Mol Sci ; 22(8)2021 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-33920085

RESUMO

Hearing loss is the most common sensory disorder with ~466 million people worldwide affected, representing about 5% of the population. A substantial portion of hearing loss is genetic. Hearing loss can either be non-syndromic, if hearing loss is the only clinical manifestation, or syndromic, if the hearing loss is accompanied by a collage of other clinical manifestations. Usher syndrome is a syndromic form of genetic hearing loss that is accompanied by impaired vision associated with retinitis pigmentosa and, in many cases, vestibular dysfunction. It is the most common cause of deaf-blindness. Currently cochlear implantation or hearing aids are the only treatments for Usher-related hearing loss. However, gene therapy has shown promise in treating Usher-related retinitis pigmentosa. Here we review how the etiologies of Usher-related hearing loss make it a good candidate for gene therapy and discuss how various forms of gene therapy could be applied to Usher-related hearing loss.


Assuntos
Orelha Interna/patologia , Terapia Genética , Perda Auditiva/terapia , Retinose Pigmentar/terapia , Síndromes de Usher/terapia , Orelha Interna/crescimento & desenvolvimento , Perda Auditiva/etiologia , Perda Auditiva/genética , Perda Auditiva/patologia , Humanos , Mutação/genética , Retinose Pigmentar/genética , Retinose Pigmentar/patologia , Síndromes de Usher/etiologia , Síndromes de Usher/genética , Síndromes de Usher/patologia
13.
Differentiation ; 101: 16-24, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29626713

RESUMO

A variety of approaches have been developed for the derivation of hepatocyte-like cells from pluripotent stem cells. Currently, most of these strategies employ step-wise differentiation approaches with recombinant growth-factors or small-molecule analogs to recapitulate developmental signaling pathways. Here, we tested the efficacy of a small-molecule based differentiation protocol for the generation of hepatocyte-like cells from human pluripotent stem cells. Quantitative gene-expression, immunohistochemical, and western blot analyses for SOX17, FOXA2, CXCR4, HNF4A, AFP, indicated the stage-specific differentiation into definitive endoderm, hepatoblast and hepatocyte-like derivatives. Furthermore, hepatocyte-like cells displayed morphological and functional features characteristic of primary hepatocytes, as indicated by the production of ALB (albumin) and α-1-antitrypsin (A1AT), as well as glycogen storage capacity by periodic acid-Schiff staining. Together, these data support that the small-molecule based hepatic differentiation protocol is a simple, reproducible, and inexpensive method to efficiently drive the differentiation of human pluripotent stem cells towards a hepatocyte-like phenotype, for downstream pharmacogenomic and regenerative medicine applications.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Piridinas/farmacologia , Pirimidinas/farmacologia , Endoderma/citologia , Endoderma/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Hepatócitos/citologia , Humanos , Fígado/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia
14.
J Neurosci ; 37(19): 4967-4981, 2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28411269

RESUMO

What pathways specify retinal ganglion cell (RGC) fate in the developing retina? Here we report on mechanisms by which a molecular pathway involving Sox4/Sox11 is required for RGC differentiation and for optic nerve formation in mice in vivo, and is sufficient to differentiate human induced pluripotent stem cells into electrophysiologically active RGCs. These data place Sox4 downstream of RE1 silencing transcription factor in regulating RGC fate, and further describe a newly identified, Sox4-regulated site for post-translational modification with small ubiquitin-related modifier (SUMOylation) in Sox11, which suppresses Sox11's nuclear localization and its ability to promote RGC differentiation, providing a mechanism for the SoxC familial compensation observed here and elsewhere in the nervous system. These data define novel regulatory mechanisms for this SoxC molecular network, and suggest pro-RGC molecular approaches for cell replacement-based therapies for glaucoma and other optic neuropathies.SIGNIFICANCE STATEMENT Glaucoma is the most common cause of blindness worldwide and, along with other optic neuropathies, is characterized by loss of retinal ganglion cells (RGCs). Unfortunately, vision and RGC loss are irreversible, and lead to bilateral blindness in ∼14% of all diagnosed patients. Differentiated and transplanted RGC-like cells derived from stem cells have the potential to replace neurons that have already been lost and thereby to restore visual function. These data uncover new mechanisms of retinal progenitor cell (RPC)-to-RGC and human stem cell-to-RGC fate specification, and take a significant step toward understanding neuronal and retinal development and ultimately cell-transplant therapy.


Assuntos
Envelhecimento/fisiologia , Redes Reguladoras de Genes/fisiologia , Células Ganglionares da Retina/fisiologia , Fatores de Transcrição SOXC/metabolismo , Ativação Transcricional/fisiologia , Vias Visuais/fisiologia , Animais , Células Cultivadas , Retroalimentação Fisiológica/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Masculino , Camundongos , Ratos Sprague-Dawley
15.
Arterioscler Thromb Vasc Biol ; 37(2): 280-290, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27908889

RESUMO

OBJECTIVE: Lineage-negative bone marrow cells (lin- BMCs) are enriched in endothelial progenitor cells and mediate vascular repair. Aging-associated senescence and apoptosis result in reduced number and functionality of lin- BMCs, impairing their prorepair capacity. The molecular mechanisms underlying lin- BMC senescence and apoptosis are poorly understood. MicroRNAs (miRNAs) regulate many important biological processes. The identification of miRNA-mRNA networks that modulate the health and functionality of lin- BMCs is a critical step in understanding the process of vascular repair. The aim of this study was to characterize the role of the miR-146a-Polo-like kinase 2 (Plk2) network in regulating lin- BMC senescence, apoptosis, and their angiogenic capability. APPROACH AND RESULTS: Transcriptome analysis in lin- BMCs isolated from young and aged wild-type and ApoE-/- (apolipoprotein E) mice showed a significant age-associated increase in miR-146a expression. In silico analysis, expression study and Luciferase reporter assay established Plk2 as a direct target of miR-146a. miR-146a overexpression in young lin- BMCs inhibited Plk2 expression, resulting in increased senescence and apoptosis, via p16Ink4a/p19Arf and p53, respectively, as well as impaired angiogenic capacity in vitro and in vivo. Conversely, suppression of miR-146a in aged lin- BMCs increased Plk2 expression and rejuvenated lin- BMCs, resulting in decreased senescence and apoptosis, leading to improved angiogenesis. CONCLUSIONS: (1) miR-146a regulates lin- BMC senescence and apoptosis by suppressing Plk2 expression that, in turn, activates p16Ink4a/p19Arf and p53 and (2) modulation of miR-146a or its target Plk2 may represent a potential therapeutic intervention to improve lin- BMC-mediated angiogenesis and vascular repair.


Assuntos
Apoptose , Células da Medula Óssea/enzimologia , Linhagem da Célula , Senescência Celular , Células Progenitoras Endoteliais/enzimologia , MicroRNAs/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Regiões 3' não Traduzidas , Fatores Etários , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Sítios de Ligação , Células da Medula Óssea/patologia , Movimento Celular , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Regulação para Baixo , Células Progenitoras Endoteliais/patologia , Genótipo , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Neovascularização Fisiológica , Fenótipo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , Transdução de Sinais , Transcriptoma , Transfecção , Proteína Supressora de Tumor p53/metabolismo
16.
Mol Cell ; 35(5): 610-25, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19748357

RESUMO

miR-24, upregulated during terminal differentiation of multiple lineages, inhibits cell-cycle progression. Antagonizing miR-24 restores postmitotic cell proliferation and enhances fibroblast proliferation, whereas overexpressing miR-24 increases the G1 compartment. The 248 mRNAs downregulated upon miR-24 overexpression are highly enriched for DNA repair and cell-cycle regulatory genes that form a direct interaction network with prominent nodes at genes that enhance (MYC, E2F2, CCNB1, and CDC2) or inhibit (p27Kip1 and VHL) cell-cycle progression. miR-24 directly regulates MYC and E2F2 and some genes that they transactivate. Enhanced proliferation from antagonizing miR-24 is abrogated by knocking down E2F2, but not MYC, and cell proliferation, inhibited by miR-24 overexpression, is rescued by miR-24-insensitive E2F2. Therefore, E2F2 is a critical miR-24 target. The E2F2 3'UTR lacks a predicted miR-24 recognition element. In fact, miR-24 regulates expression of E2F2, MYC, AURKB, CCNA2, CDC2, CDK4, and FEN1 by recognizing seedless but highly complementary sequences.


Assuntos
Regiões 3' não Traduzidas , Ciclo Celular/genética , Proliferação de Células , Fator de Transcrição E2F2/genética , Genes cdc , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Sequências Reguladoras de Ácido Nucleico , Sequência de Bases , Sítios de Ligação , Diferenciação Celular/genética , Reparo do DNA , Bases de Dados Genéticas , Regulação para Baixo , Eritrócitos/metabolismo , Fibroblastos/metabolismo , Redes Reguladoras de Genes , Células HL-60 , Humanos , Células K562 , Macrófagos/metabolismo , Megacariócitos/metabolismo , Dados de Sequência Molecular , Interferência de RNA , RNA Mensageiro/metabolismo , Ativação Transcricional
17.
Mol Cell ; 31(1): 33-46, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18614045

RESUMO

The histone H2A variant H2AX is rapidly phosphorylated in response to DNA double-stranded breaks to produce gamma-H2AX. gamma-H2AX stabilizes cell-cycle checkpoint proteins and DNA repair factors at the break site. We previously found that the protein phosphatase PP2A is required to resolve gamma-H2AX foci and complete DNA repair after exogenous DNA damage. Here we describe a three-protein PP4 phosphatase complex in mammalian cells, containing PP4C, PP4R2, and PP4R3beta, that specifically dephosphorylates ATR-mediated gamma-H2AX generated during DNA replication. PP4 efficiently dephosphorylates gamma-H2AX within mononucleosomes in vitro and does not directly alter ATR or checkpoint kinase activity, suggesting that PP4 acts directly on gamma-H2AX in cells. When the PP4 complex is silenced, repair of DNA replication-mediated breaks is inefficient, and cells are hypersensitive to DNA replication inhibitors, but not radiomimetic drugs. Therefore, gamma-H2AX elimination at DNA damage foci is required for DNA damage repair, but accomplishing this task involves distinct phosphatases with potentially overlapping roles.


Assuntos
Replicação do DNA , Histonas/metabolismo , Complexos Multiproteicos/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Sequência de Aminoácidos , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , Linhagem Celular , Cromatina/metabolismo , Dano ao DNA , Inativação Gênica , Humanos , Dados de Sequência Molecular , Fosfoproteínas Fosfatases/química , Fosforilação , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Subunidades Proteicas/metabolismo , Transporte Proteico , Frações Subcelulares/metabolismo
18.
Mol Cell Neurosci ; 68: 244-57, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26284979

RESUMO

BACKGROUND: GABAergic synaptic transmission is known to play a critical role in the assembly of neuronal circuits during development and is responsible for maintaining the balance between excitatory and inhibitory signaling in the brain during maturation into adulthood. Importantly, defects in GABAergic neuronal function and signaling have been linked to a number of neurological diseases, including autism spectrum disorders, schizophrenia, and epilepsy. With patient-specific induced pluripotent stem cell (iPSC)-based models of neurological disease, it is now possible to investigate the disease mechanisms that underlie deficits in GABAergic function in affected human neurons. To that end, tools that enable the labeling and purification of viable GABAergic neurons from human pluripotent stem cells would be of great value. RESULTS: To address the need for tools that facilitate the identification and isolation of viable GABAergic neurons from the in vitro differentiation of iPSC lines, a cell type-specific promoter-driven fluorescent reporter construct was developed that utilizes the human vesicular GABA transporter (hVGAT) promoter to drive the expression of mCherry specifically in VGAT-expressing neurons. The transduction of iPSC-derived forebrain neuronal cultures with the hVGAT promoter-mCherry lentiviral reporter construct specifically labeled GABAergic neurons. Immunocytochemical analysis of hVGAT-mCherry expression cells showed significant co-labeling with the GABAergic neuronal markers for endogenous VGAT, GABA, and GAD67. Expression of mCherry from the VGAT promoter showed expression in several cortical interneuron subtypes to similar levels. In addition, an effective and reproducible protocol was developed to facilitate the fluorescent activated cell sorting (FACS)-mediated purification of high yields of viable VGAT-positive cells. CONCLUSIONS: These studies demonstrate the utility of the hVGAT-mCherry reporter construct as an effective tool for studying GABAergic neurons differentiated in vitro from human pluripotent stem cells. This approach could provide a means of obtaining large quantities of viable GABAergic neurons derived from disease-specific hiPSCs that could be used for functional assays or high-throughput screening of small molecule libraries.


Assuntos
Neurônios GABAérgicos/metabolismo , Proteínas Luminescentes/metabolismo , Células-Tronco Pluripotentes/fisiologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Calbindina 2/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular , Células Cultivadas , Citometria de Fluxo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Glutamato Descarboxilase/metabolismo , Humanos , Proteínas Luminescentes/genética , Mutação/genética , Fator de Crescimento Neural/farmacologia , Parvalbuminas/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Prosencéfalo/citologia , Somatostatina/metabolismo , Sinapsinas/metabolismo , Transfecção , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteína Vermelha Fluorescente
19.
Circ Res ; 112(1): 152-64, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23072816

RESUMO

RATIONALE: Endothelial progenitor cells (EPCs) contribute to the regeneration of endothelium. Aging-associated senescence results in reduced number and function of EPCs, potentially contributing to increased cardiac risk, reduced angiogenic capacity, and impaired cardiac repair effectiveness. The mechanisms underlying EPC senescence are unknown. Increasing evidence supports the role of microRNAs in regulating cellular senescence. OBJECTIVE: We aimed to determine whether microRNAs regulated EPC senescence and, if so, what the underlying mechanisms are. METHODS AND RESULTS: To map the microRNA/gene expression signatures of EPC senescence, we performed microRNA profiling and microarray analysis in lineage-negative bone marrow cells from young and aged wild-type and apolipoprotein E-deficient mice. We identified 2 microRNAs, microRNA-10A* (miR-10A*), and miR-21, and their common target gene Hmga2 as critical regulators for EPC senescence. Overexpression of miR-10A* and miR-21 in young EPCs suppressed Hmga2 expression, caused EPC senescence, as evidenced by senescence-associated ß-galactosidase upregulation, decreased self-renewal potential, increased p16(Ink4a)/p19(Arf) expression, and resulted in impaired EPC angiogenesis in vitro and in vivo, resembling EPCs derived from aged mice. In contrast, suppression of miR-10A* and miR-21 in aged EPCs increased Hmga2 expression, rejuvenated EPCs, resulting in decreased senescence-associated ß-galactosidase expression, increased self-renewal potential, decreased p16(Ink4a)/p19(Arf) expression, and improved EPC angiogenesis in vitro and in vivo. Importantly, these phenotypic changes were rescued by miRNA-resistant Hmga2 cDNA overexpression. CONCLUSIONS: miR-10A* and miR-21 regulate EPC senescence via suppressing Hmga2 expression and modulation of microRNAs may represent a potential therapeutic intervention in improving EPC-mediated angiogenesis and vascular repair.


Assuntos
Senescência Celular , Células Endoteliais/metabolismo , Proteína HMGB3/metabolismo , MicroRNAs/metabolismo , Células-Tronco/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Proliferação de Células , Células Cultivadas , Senescência Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Perfilação da Expressão Gênica/métodos , Genótipo , Proteína HMGB3/genética , Membro Posterior , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Transfecção , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
20.
Res Sq ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38947059

RESUMO

Developmental anomalies of the hearing organ, the cochlea, are diagnosed in approximately one-fourth of individuals with congenital deafness. Most patients with cochlear malformations remain etiologically undiagnosed due to insufficient knowledge about underlying genes or the inability to make conclusive interpretations of identified genetic variants. We used exome sequencing for genetic evaluation of hearing loss associated with cochlear malformations in three probands from unrelated families. We subsequently generated monoclonal induced pluripotent stem cell (iPSC) lines, bearing patient-specific knockins and knockouts using CRISPR/Cas9 to assess pathogenicity of candidate variants. We detected FGF3 (p.Arg165Gly) and GREB1L (p.Cys186Arg), variants of uncertain significance in two recognized genes for deafness, and PBXIP1 (p.Trp574*) in a candidate gene. Upon differentiation of iPSCs towards inner ear organoids, we observed significant developmental aberrations in knockout lines compared to their isogenic controls. Patient-specific single nucleotide variants (SNVs) showed similar abnormalities as the knockout lines, functionally supporting their causality in the observed phenotype. Therefore, we present human inner ear organoids as a tool to rapidly validate the pathogenicity of DNA variants associated with cochlear malformations.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA