Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Genomics ; 53(4): 160-171, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33719581

RESUMO

During pregnancy, the uterine spiral arteries undergo major vascular remodeling to ensure sufficient uteroplacental perfusion to support the fetus. In pregnancies complicated by hypertensive disorders, this remodeling is deficient leading to impaired uteroplacental blood flow and poor maternal and fetal outcomes. The underlying genetic mechanisms for failed vascular remodeling are not fully understood. This study aimed to examine the early-pregnancy-associated gene changes in the uterine arteries of spontaneously hypertensive stroke-prone rats (SHRSP) compared with their normotensive counterparts, Wistar-Kyoto rats (WKY). Uterine arteries from gestational day 6.5 WKY and SHRSP were processed for RNA-sequencing, along with virgin, age-matched controls for each strain. Gene expression changes were identified and biological pathways were implicated and interpretated using ingenuity pathway analysis (IPA). This study found that WKY uterine arteries from early pregnancy exhibit a gene expression pattern that is suggestive of a pregnancy-dependent reduction in Ca2+ handling and renin-angiotensin-aldosterone system (RAAS) components and an increase in ATP production. In contrast, the expression pattern of pregnant SHRSP uterine arteries was dominated by an elevated immune response and increased production of reactive oxygen species (ROS) and downstream effectors of the RAAS. These results suggest that in a rat model, hypertension during pregnancy impacts uterine artery gene expression patterns as early as the first week of pregnancy. The pathway changes involved may underlie or contribute to the adverse vascular remodeling and resultant placental ischemia and systemic vascular dysfunction observed in SHRSP in late gestation.


Assuntos
Hipertensão , Acidente Vascular Cerebral , Animais , Feminino , Placenta/metabolismo , Gravidez , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Acidente Vascular Cerebral/etiologia , Transcriptoma/genética , Artéria Uterina/metabolismo
2.
Clin Sci (Lond) ; 134(8): 1001-1025, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32337535

RESUMO

Placental microRNAs (miRNAs) regulate the placental transcriptome and play a pathological role in preeclampsia (PE), a hypertensive disorder of pregnancy. Three PE rodent model studies explored the role of placental miRNAs, miR-210, miR-126, and miR-148/152 respectively, by examining expression of the miRNAs, their inducers, and potential gene targets. This review evaluates the role of miR-210, miR-126, and miR-148/152 in PE by comparing findings from the three rodent model studies with in vitro studies, other animal models, and preeclamptic patients to provide comprehensive insight into genetic components and pathological processes in the placenta contributing to PE. The majority of studies demonstrate miR-210 is upregulated in PE in part driven by HIF-1α and NF-κBp50, stimulated by hypoxia and/or immune-mediated processes. Elevated miR-210 may contribute to PE via inhibiting anti-inflammatory Th2-cytokines. Studies report an up- and downregulation of miR-126, arguably reflecting differences in expression between cell types and its multifunctional capacity. MiR-126 may play a pro-angiogenic role by mediating the PI3K-Akt pathway. Most studies report miR-148/152 family members are upregulated in PE. Evidence suggests they may inhibit DNA methylation of genes involved in metabolic and inflammatory pathways. Given the genetic heterogeneity of PE, it is unlikely that a single placental miRNA is a suitable therapeutic target for all patients. Investigating miRNAs in PE subtypes in patients and animal models may represent a more appropriate approach going forward. Developing methods for targeting placental miRNAs and specific placental cell types remains crucial for research seeking to target placental miRNAs as a novel treatment for PE.


Assuntos
MicroRNAs/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , MicroRNAs/genética , Pré-Eclâmpsia/genética , Gravidez
3.
Circulation ; 137(1): 57-70, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29030345

RESUMO

BACKGROUND: Myocardial infarction (MI) is a leading cause of heart failure and death worldwide. Preservation of contractile function and protection against adverse changes in ventricular architecture (cardiac remodeling) are key factors to limiting progression of this condition to heart failure. Consequently, new therapeutic targets are urgently required to achieve this aim. Expression of the Runx1 transcription factor is increased in adult cardiomyocytes after MI; however, the functional role of Runx1 in the heart is unknown. METHODS: To address this question, we have generated a novel tamoxifen-inducible cardiomyocyte-specific Runx1-deficient mouse. Mice were subjected to MI by means of coronary artery ligation. Cardiac remodeling and contractile function were assessed extensively at the whole-heart, cardiomyocyte, and molecular levels. RESULTS: Runx1-deficient mice were protected against adverse cardiac remodeling after MI, maintaining ventricular wall thickness and contractile function. Furthermore, these mice lacked eccentric hypertrophy, and their cardiomyocytes exhibited markedly improved calcium handling. At the mechanistic level, these effects were achieved through increased phosphorylation of phospholamban by protein kinase A and relief of sarco/endoplasmic reticulum Ca2+-ATPase inhibition. Enhanced sarco/endoplasmic reticulum Ca2+-ATPase activity in Runx1-deficient mice increased sarcoplasmic reticulum calcium content and sarcoplasmic reticulum-mediated calcium release, preserving cardiomyocyte contraction after MI. CONCLUSIONS: Our data identified Runx1 as a novel therapeutic target with translational potential to counteract the effects of adverse cardiac remodeling, thereby improving survival and quality of life among patients with MI.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Função Ventricular Esquerda , Remodelação Ventricular , Animais , Sinalização do Cálcio , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Fosforilação , Coelhos , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/patologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Fatores de Tempo
4.
Physiol Genomics ; 50(11): 964-972, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30216136

RESUMO

Previously, our comprehensive cardiovascular characterization study validated Uromodulin as a blood pressure gene. Uromodulin is a glycoprotein exclusively synthesized at the thick ascending limb of the loop of Henle and is encoded by the Umod gene. Umod-/- mice have significantly lower blood pressure than Umod+/+ mice, are resistant to salt-induced changes in blood pressure, and show a leftward shift in pressure-natriuresis curves reflecting changes of sodium reabsorption. Salt stress triggers transcription factors and genes that alter renal sodium reabsorption. To date there are no studies on renal transcriptome responses to salt stress. Here we aimed use RNA-Seq to delineate salt stress pathways in tubules isolated from Umod+/+ mice (a model of sodium retention) and Umod-/- mice (a model of sodium depletion) ± 300 mosmol sodium chloride ( n = 3 per group). In response to salt stress, the tubules of Umod+/+ mice displayed an upregulation of heat shock transcripts. The greatest changes occurred in the expression of: Hspa1a (Log2 fold change 4.35, P = 2.48 e-12) and Hspa1b (Log2 fold change 4.05, P = 2.48 e-12). This response was absent in tubules of Umod-/- mice. Interestingly, seven of the genes discordantly expressed in the Umod-/- tubules were electrolyte transporters. Our results are the first to show that salt stress in renal tubules alters the transcriptome, increasing the expression of heat shock genes. This direction of effect in Umod+/+ tubules suggest the difference is due to the presence of Umod facilitating greater sodium entry into the tubule cell reflecting a specific response to salt stress.


Assuntos
Resposta ao Choque Térmico/genética , Túbulos Renais/fisiologia , Estresse Salino/genética , Uromodulina/genética , Animais , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Alça do Néfron/fisiologia , Masculino , Camundongos Mutantes , Regulação para Cima
5.
Clin Sci (Lond) ; 132(12): 1315-1328, 2018 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-29632138

RESUMO

BACKGROUND: The effect of salt on cerebral small vessel disease (SVD) is poorly understood. We assessed the effect of dietary salt on cerebral tissue of the stroke-prone spontaneously hypertensive rat (SHRSP) - a relevant model of sporadic SVD - at both the gene and protein level. Methods: Brains from 21-week-old SHRSP and Wistar-Kyoto rats, half additionally salt-loaded (via a 3-week regime of 1% NaCl in drinking water), were split into two hemispheres and sectioned coronally - one hemisphere for mRNA microarray and qRT-PCR, the other for immunohistochemistry using a panel of antibodies targeting components of the neurovascular unit. Results: We observed differences in gene and protein expression affecting the acute phase pathway and oxidative stress (ALB, AMBP, APOH, AHSG and LOC100129193, up-regulated in salt-loaded WKY versus WKY, >2-fold), active microglia (increased Iba-1 protein expression in salt-loaded SHRSP versus salt-loaded WKY, p<0.05), vascular structure (ACTB and CTNNB, up-regulated in salt-loaded SHRSP versus SHRSP, >3-fold; CLDN-11, VEGF and VGF down-regulated >2-fold in salt-loaded SHRSP versus SHRSP) and myelin integrity (MBP down-regulated in salt loaded WKY rats versus WKY, >2.5-fold). Changes of salt-loading were more pronounced in SHRSP and occurred without an increase in blood pressure in WKY rats. CONCLUSION: Salt exposure induced changes in gene and protein expression in an experimental model of SVD and its parent rat strain in multiple pathways involving components of the glio-vascular unit. Further studies in pertinent experimental models at different ages would help clarify the short- and long-term effect of dietary salt in SVD.


Assuntos
Encéfalo/metabolismo , Doenças de Pequenos Vasos Cerebrais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Cloreto de Sódio na Dieta/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Estresse Oxidativo , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Regulação para Cima/efeitos dos fármacos
6.
Physiol Genomics ; 49(3): 193-200, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28130428

RESUMO

Preeclampsia is a multisystem disease that significantly contributes to maternal and fetal morbidity and mortality. In this study, we used a non-biased microarray approach to identify dysregulated genes in maternal whole blood samples which may be associated with the development of preeclampsia. Whole blood samples were obtained at 28 wk of gestation from 5 women who later developed preeclampsia (cases) and 10 matched women with normotensive pregnancies (controls). Placenta samples were obtained from an independent cohort of 19 women with preeclampsia matched with 19 women with normotensive pregnancies. We studied gene expression profiles using Illumina microarray in blood and validated changes in gene expression in whole blood and placenta tissue by qPCR. We found a transcriptional profile differentiating cases from controls; 336 genes were significantly dysregulated in blood from women who developed preeclampsia. Functional annotation of microarray results indicated that most of the genes found to be dysregulated were involved in inflammatory pathways. While general trends were preserved, only HLA-A was validated in whole blood samples from cases using qPCR (2.30- ± 0.9-fold change) whereas in placental tissue HLA-DRB1 expression was found to be significantly increased in samples from women with preeclampsia (5.88- ± 2.24-fold change). We have identified that HLA-A is upregulated in the circulation of women who went on to develop preeclampsia. In placenta of women with preeclampsia we identified that HLA-DRB1 is upregulated. Our data provide further evidence for involvement of the HLA gene family in the pathogenesis of preeclampsia.


Assuntos
Regulação da Expressão Gênica , Antígenos HLA/genética , Placenta/metabolismo , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/genética , Adulto , Feminino , Antígenos HLA/metabolismo , Humanos , Gravidez , Transcriptoma , Regulação para Cima/genética
7.
Circulation ; 133(21): 2050-65, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27052414

RESUMO

BACKGROUND: Phenotypic switching of vascular smooth muscle cells from a contractile to a synthetic state is implicated in diverse vascular pathologies, including atherogenesis, plaque stabilization, and neointimal hyperplasia. However, very little is known about the role of long noncoding RNA (lncRNA) during this process. Here, we investigated a role for lncRNAs in vascular smooth muscle cell biology and pathology. METHODS AND RESULTS: Using RNA sequencing, we identified >300 lncRNAs whose expression was altered in human saphenous vein vascular smooth muscle cells following stimulation with interleukin-1α and platelet-derived growth factor. We focused on a novel lncRNA (Ensembl: RP11-94A24.1), which we termed smooth muscle-induced lncRNA enhances replication (SMILR). Following stimulation, SMILR expression was increased in both the nucleus and cytoplasm, and was detected in conditioned media. Furthermore, knockdown of SMILR markedly reduced cell proliferation. Mechanistically, we noted that expression of genes proximal to SMILR was also altered by interleukin-1α/platelet-derived growth factor treatment, and HAS2 expression was reduced by SMILR knockdown. In human samples, we observed increased expression of SMILR in unstable atherosclerotic plaques and detected increased levels in plasma from patients with high plasma C-reactive protein. CONCLUSIONS: These results identify SMILR as a driver of vascular smooth muscle cell proliferation and suggest that modulation of SMILR may be a novel therapeutic strategy to reduce vascular pathologies.


Assuntos
Proliferação de Células/fisiologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , RNA Longo não Codificante/fisiologia , Proteínas de Caenorhabditis elegans , Células Cultivadas , Técnicas de Silenciamento de Genes , Humanos , Músculo Liso Vascular/citologia , Veia Safena/citologia , Veia Safena/fisiologia
8.
Physiol Genomics ; 48(3): 202-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26757800

RESUMO

Recombinant human erythropoietin (rHuEPO) is frequently abused by athletes as a performance-enhancing drug, despite being prohibited by the World Anti-Doping Agency. Although the methods to detect blood doping, including rHuEPO injections, have improved in recent years, they remain imperfect. In a proof-of-principle study, we identified, replicated, and validated the whole blood transcriptional signature of rHuEPO in endurance-trained Caucasian males at sea level (n = 18) and Kenyan endurance runners at moderate altitude (n = 20), all of whom received rHuEPO injections for 4 wk. Transcriptional profiling shows that hundreds of transcripts were altered by rHuEPO in both cohorts. The main regulated expression pattern, observed in all participants, was characterized by a "rebound" effect with a profound upregulation during rHuEPO and a subsequent downregulation up to 4 wk postadministration. The functions of the identified genes were mainly related to the functional and structural properties of the red blood cell. Of the genes identified to be differentially expressed during and post-rHuEPO, we further confirmed a whole blood 34-transcript signature that can distinguish between samples collected pre-, during, and post-rHuEPO administration. By providing biomarkers that can reveal rHuEPO use, our findings represent an advance in the development of new methods for the detection of blood doping.


Assuntos
Dopagem Esportivo/prevenção & controle , Eritropoetina/sangue , Eritropoetina/genética , Proteínas Recombinantes/sangue , Proteínas Recombinantes/genética , Adulto , Eritropoetina/administração & dosagem , Eritropoetina/biossíntese , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/biossíntese , Transcrição Gênica
9.
Neuropathol Appl Neurobiol ; 40(7): 855-72, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24417612

RESUMO

AIMS: Cerebral small vessel disease (SVD) causes a fifth of all strokes plus diffuse brain damage leading to cognitive decline, physical disabilities and dementia. The aetiology and pathogenesis of SVD are unknown, but largely attributed to hypertension or microatheroma. METHODS: We used the spontaneously hypertensive stroke-prone rat (SHRSP), the closest spontaneous experimental model of human SVD, and age-matched control rats kept under identical, non-salt-loaded conditions, to perform a blinded analysis of mRNA microarray, qRT-PCR and pathway analysis in two brain regions (frontal and mid-coronal) commonly affected by SVD in the SHRSP at age five, 16 and 21 weeks. RESULTS: We found gene expression abnormalities, with fold changes ranging from 2.5 to 59 for the 10 most differentially expressed genes, related to endothelial tight junctions (reduced), nitric oxide bioavailability (reduced), myelination (impaired), glial and microglial activity (increased), matrix proteins (impaired), vascular reactivity (impaired) and albumin (reduced), consistent with protein expression defects in the same rats. All were present at age 5 weeks thus predating blood pressure elevation. 'Neurological' and 'inflammatory' pathways were more affected than 'vascular' functional pathways. CONCLUSIONS: This set of defects, although individually modest, when acting in combination could explain the SHRSP's susceptibility to microvascular and brain injury, compared with control rats. Similar combined, individually modest, but multiple neurovascular unit defects, could explain susceptibility to spontaneous human SVD.


Assuntos
Encéfalo/metabolismo , Doenças de Pequenos Vasos Cerebrais/complicações , Doenças de Pequenos Vasos Cerebrais/genética , Animais , Tecido Conjuntivo/metabolismo , Modelos Animais de Doenças , Encefalite/complicações , Encefalite/genética , Expressão Gênica , Humanos , Masculino , Doenças do Sistema Nervoso/complicações , Doenças do Sistema Nervoso/genética , Análise Serial de Proteínas , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR
10.
Cardiovasc Res ; 119(16): 2663-2671, 2023 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-37433039

RESUMO

AIMS: Myocardial infarction (MI) is a major cause of death worldwide. Effective treatments are required to improve recovery of cardiac function following MI, with the aim of improving patient outcomes and preventing progression to heart failure. The perfused but hypocontractile region bordering an infarct is functionally distinct from the remote surviving myocardium and is a determinant of adverse remodelling and cardiac contractility. Expression of the transcription factor RUNX1 is increased in the border zone 1-day after MI, suggesting potential for targeted therapeutic intervention. OBJECTIVE: This study sought to investigate whether an increase in RUNX1 in the border zone can be therapeutically targeted to preserve contractility following MI. METHODS AND RESULTS: In this work we demonstrate that Runx1 drives reductions in cardiomyocyte contractility, calcium handling, mitochondrial density, and expression of genes important for oxidative phosphorylation. Both tamoxifen-inducible Runx1-deficient and essential co-factor common ß subunit (Cbfß)-deficient cardiomyocyte-specific mouse models demonstrated that antagonizing RUNX1 function preserves the expression of genes important for oxidative phosphorylation following MI. Antagonizing RUNX1 expression via short-hairpin RNA interference preserved contractile function following MI. Equivalent effects were obtained with a small molecule inhibitor (Ro5-3335) that reduces RUNX1 function by blocking its interaction with CBFß. CONCLUSIONS: Our results confirm the translational potential of RUNX1 as a novel therapeutic target in MI, with wider opportunities for use across a range of cardiac diseases where RUNX1 drives adverse cardiac remodelling.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Animais , Camundongos , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Insuficiência Cardíaca/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/prevenção & controle , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Remodelação Ventricular
11.
J Neurosci ; 31(37): 13028-38, 2011 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-21917786

RESUMO

Astrocytes undergo major phenotypic changes in response to injury and disease that directly influence repair in the CNS, but the mechanisms involved are poorly understood. Previously, we have shown that neurosphere-derived rat astrocytes plated on poly-L-lysine (PLL-astrocytes) support myelination in dissociated rat spinal cord cultures (myelinating cultures). It is hypothesized that astrocyte reactivity can affect myelination, so we have exploited this culture system to ascertain how two distinct astrocyte phenotypes influence myelination. Astrocytes plated on tenascin C (TnC-astrocytes), a method to induce quiescence, resulted in less myelinated fibers in the myelinating cultures when compared with PLL-astrocytes. In contrast, treatment of myelinating cultures plated on PLL-astrocytes with ciliary neurotrophic factor (CNTF), a cytokine known to induce an activated astrocyte phenotype, promoted myelination. CNTF could also reverse the effect of quiescent astrocytes on myelination. A combination of microarray gene expression analysis and quantitative real-time PCR identified CXCL10 as a potential candidate for the reduction in myelination in cultures on TnC-astrocytes. The effect of TnC-astrocytes on myelination was eliminated by neutralizing CXCL10 antibodies. Conversely, CXCL10 protein inhibited myelination on PLL-astrocytes. Furthermore, CXCL10 treatment of purified oligodendrocyte precursor cells did not affect proliferation, differentiation, or process extension compared with untreated controls, suggesting a role in glial/axonal ensheathment. These data demonstrate a direct correlation of astrocyte phenotypes with their ability to support myelination. This observation has important implications with respect to the development of therapeutic strategies to promote CNS remyelination in demyelinating diseases.


Assuntos
Astrócitos/metabolismo , Quimiocina CXCL10/fisiologia , Fibras Nervosas Mielinizadas/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Células Cultivadas , Fator Neurotrófico Ciliar/fisiologia , Meios de Cultura , Feminino , Masculino , Fibras Nervosas Mielinizadas/fisiologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Fenótipo , Polilisina/fisiologia , Análise Serial de Proteínas/métodos , Ratos , Ratos Sprague-Dawley
12.
Am J Pathol ; 179(2): 661-72, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21704009

RESUMO

Transforming growth factor (TGF)-ß is one of the main fibrogenic cytokines that drives the pathophysiology of progressive renal scarring. MicroRNAs (miRNAs) are endogenous non-coding RNAs that post-transcriptionally regulate gene expression. We examined the role of TGF-ß-induced expression of miR-21, miRNAs in cell culture models and miRNA expression in relevant models of renal disease. In vitro, TGF-ß changed expression of miR-21, miR-214, and miR-145 in rat mesangial cells (CRL-2753) and miR-214, miR-21, miR-30c, miR-200b, and miR-200c during induction of epithelial-mesenchymal transition in rat tubular epithelial cells (NRK52E). miR-214 expression was robustly modulated in both cell types, whereas in tubular epithelial cells miR-21 was increased and miR-200b and miR-200c were decreased by 58% and 48%, respectively, in response to TGF-ß. TGF-ß receptor-1 was found to be a target of miR-200b/c and was down-regulated after overexpression of miR-200c. To assess the differential expression of these miRNAs in vivo, we used the anti-Thy1.1 mesangial glomerulonephritis model and the unilateral ureteral obstruction model in which TGF-ß plays a role and also a genetic model of hypertension, the stroke-prone spontaneously hypertensive rat with and without salt loading. The expressions of miR-214 and miR-21 were significantly increased in all in vivo models, showing a possible miRNA signature of renal damage despite differing causes.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Modelos Animais de Doenças , Glomerulonefrite/metabolismo , Hipertensão/patologia , Rim/lesões , Rim/metabolismo , Glomérulos Renais/metabolismo , Túbulos Renais/metabolismo , Masculino , Ratos , Ratos Endogâmicos WKY , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Ureter/patologia
13.
Nucleic Acids Res ; 38(20): 6831-40, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20571087

RESUMO

This article describes and illustrates a novel method of microarray data analysis that couples model-based clustering and binary classification to form clusters of `response-relevant' genes; that is, genes that are informative when discriminating between the different values of the response. Predictions are subsequently made using an appropriate statistical summary of each gene cluster, which we call the `meta-covariate' representation of the cluster, in a probit regression model. We first illustrate this method by analysing a leukaemia expression dataset, before focusing closely on the meta-covariate analysis of a renal gene expression dataset in a rat model of salt-sensitive hypertension. We explore the biological insights provided by our analysis of these data. In particular, we identify a highly influential cluster of 13 genes--including three transcription factors (Arntl, Bhlhe41 and Npas2)-that is implicated as being protective against hypertension in response to increased dietary sodium. Functional and canonical pathway analysis of this cluster using Ingenuity Pathway Analysis implicated transcriptional activation and circadian rhythm signalling, respectively. Although we illustrate our method using only expression data, the method is applicable to any high-dimensional datasets. Expression data are available at ArrayExpress (accession number E-MEXP-2514) and code is available at http://www.dcs.gla.ac.uk/inference/metacovariateanalysis/.


Assuntos
Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Animais , Ritmo Circadiano/genética , Análise por Conglomerados , Redes Reguladoras de Genes , Humanos , Hipertensão/genética , Hipertensão/metabolismo , Rim/metabolismo , Leucemia/genética , Leucemia/metabolismo , Ratos , Análise de Regressão
14.
Hypertension ; 79(11): 2419-2429, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36378920

RESUMO

The exclusive expression of uromodulin in the kidneys has made it an intriguing protein in kidney and cardiovascular research. Genome-wide association studies discovered variants of uromodulin that are associated with chronic kidney diseases and hypertension. Urinary and circulating uromodulin levels reflect kidney and cardiovascular health as well as overall mortality. More recently, Mendelian randomization studies have shown that genetically driven levels of uromodulin have a causal and adverse effect on kidney function. On a mechanistic level, salt sensitivity is an important factor in the pathophysiology of hypertension, and uromodulin is involved in salt reabsorption via the NKCC2 (Na+-K+-2Cl- cotransporter) on epithelial cells of the ascending limb of loop of Henle. In this review, we provide an overview of the multifaceted physiology and pathophysiology of uromodulin including recent advances in its genetics; cellular trafficking; and mechanistic and clinical studies undertaken to understand the complex relationship between uromodulin, blood pressure, and kidney function. We focus on tubular sodium reabsorption as one of the best understood and pathophysiologically and clinically most important roles of uromodulin, which can lead to therapeutic interventions.


Assuntos
Estudo de Associação Genômica Ampla , Hipertensão , Humanos , Uromodulina/genética , Uromodulina/metabolismo , Hipertensão/genética , Hipertensão/metabolismo , Pressão Sanguínea/fisiologia , Rim/metabolismo , Cloreto de Sódio na Dieta/efeitos adversos , Cloreto de Sódio na Dieta/metabolismo , Cloreto de Sódio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/genética
15.
Biochim Biophys Acta ; 1802(12): 1299-308, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20035862

RESUMO

Essential hypertension affects 20 to 30% of the population worldwide and contributes significantly to cardiovascular mortality and morbidity. Heridability of blood pressure is around 15 to 40% but there are also substantial environmental factors affecting blood pressure variability. It is assumed that blood pressure is under the control of a large number of genes each of which has only relatively mild effects. It has therefore been difficult to discover the genes that contribute to blood pressure variation using traditional approaches including candidate gene studies and linkage studies. Animal models of hypertension, particularly in the rat, have led to the discovery of quantitative trait loci harbouring one or several hypertension related genes, but translation of these findings into human essential hypertension remains challenging. Recent development of genotyping technology made large scale genome-wide association studies possible. This approach and the study of monogenic forms of hypertension has led to the discovery of novel and robust candidate genes for human essential hypertension, many of which require functional analysis in experimental models.


Assuntos
Modelos Animais de Doenças , Hipertensão/genética , Animais , Estudo de Associação Genômica Ampla , Humanos , Hipertensão/mortalidade , Hipertensão/fisiopatologia , Ratos
16.
J Mol Cell Cardiol ; 48(6): 1121-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20138888

RESUMO

The structural integrity of cardiac cells is maintained by the Ca(2+)-dependent homophilic cell-cell adhesion of cadherins. N-cadherin is responsible for this adhesion under normal physiological conditions. The role of cadherins in adverse cardiac pathology is less clear. We studied the hearts of the stroke-prone spontaneously hypertensive (SHRSP) rat as a genetic model of cardiac hypertrophy and compared them to Wistar-Kyoto control animals. Western blotting of protein homogenates from 12-week old SHRSP animals indicated that similar levels of beta, gamma-, and alpha-catenin and T, N and R-cadherin were expressed in the control and SHRSP animals. However, dramatically higher levels of E-cadherin were detected in SHRSP animals compared to controls at 6, 12 and 18 weeks of age. This was confirmed by quantitative Taqman PCR and immunohistochemistry. E-cadherin was located at the intercalated disc of the myocytes in co-localisation with connexin 43. Adenoviral overexpression of E-cadherin in rat H9c2 cells and primary rabbit myocytes resulted in a significant reduction in myocyte cell diameter and breadth. E-cadherin overexpression resulted in re-localisation of beta-catenin to the cell surface particularly to cell-cell junctions. Subsequent immunohistochemistry of the hearts of WKY and SHRSP animals also revealed increased levels of beta-catenin in the intercalated disc in the SHRSP compared to WKY. Therefore, remodelling of the intercalated disc in the hearts of SHRSP animals may contribute to the altered function observed in these animals.


Assuntos
Caderinas/metabolismo , Regulação da Expressão Gênica , Hipertensão/patologia , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/patologia , Animais , Cardiomegalia/patologia , Adesão Celular , Hipertensão/metabolismo , Imuno-Histoquímica/métodos , Miócitos Cardíacos/metabolismo , Coelhos , Ratos , Ratos Endogâmicos WKY , beta Catenina/metabolismo
17.
Clin Sci (Lond) ; 119(8): 335-43, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20528768

RESUMO

Owing to the dynamic nature of the transcriptome, gene expression profiling is a promising tool for discovery of disease-related genes and biological pathways. In the present study, we examined gene expression in whole blood of 12 patients with CAD (coronary artery disease) and 12 healthy control subjects. Furthermore, ten patients with CAD underwent whole-blood gene expression analysis before and after the completion of a cardiac rehabilitation programme following surgical coronary revascularization. mRNA and miRNA (microRNA) were isolated for expression profiling. Gene expression analysis identified 365 differentially expressed genes in patients with CAD compared with healthy controls (175 up- and 190 down-regulated in CAD), and 645 in CAD rehabilitation patients (196 up- and 449 down-regulated post-rehabilitation). Biological pathway analysis identified a number of canonical pathways, including oxidative phosphorylation and mitochondrial function, as being significantly and consistently modulated across the groups. Analysis of miRNA expression revealed a number of differentially expressed miRNAs, including hsa-miR-140-3p (control compared with CAD, P=0.017), hsa-miR-182 (control compared with CAD, P=0.093), hsa-miR-92a and hsa-miR-92b (post- compared with pre-exercise, P<0.01). Global analysis of predicted miRNA targets found significantly reduced expression of genes with target regions compared with those without: hsa-miR-140-3p (P=0.002), hsa-miR-182 (P=0.001), hsa-miR-92a and hsa-miR-92b (P=2.2x10-16). In conclusion, using whole blood as a 'surrogate tissue' in patients with CAD, we have identified differentially expressed miRNAs, differentially regulated genes and modulated pathways which warrant further investigation in the setting of cardiovascular function. This approach may represent a novel non-invasive strategy to unravel potentially modifiable pathways and possible therapeutic targets in cardiovascular disease.


Assuntos
Doença da Artéria Coronariana/genética , Perfilação da Expressão Gênica/métodos , Idoso , Estudos de Casos e Controles , Ponte de Artéria Coronária , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/cirurgia , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Mitocôndrias Cardíacas/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fosforilação Oxidativa , Período Pós-Operatório , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transdução de Sinais/genética
18.
Trends Endocrinol Metab ; 19(9): 309-16, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18819818

RESUMO

Recent advances in genotyping technology and insights into disease mechanisms have increased interest in the genetics of cardiovascular disease. Several candidate genes involved in cardiovascular diseases were identified from studies using animal models, and the translation of these findings to human disease is an exciting challenge. There is a trend towards large-scale genome-wide association studies that are subject to strict quality criteria with regard to both genotyping and phenotyping. Here, we review some of the strategies that have been developed to translate findings from experimental models to human disease and outline the need for optimizing global approaches to analyze such results. Findings from ongoing studies are interpreted in the context of disease pathways instead of the more traditional focus on single genetic variants.


Assuntos
Doenças Cardiovasculares/genética , Predisposição Genética para Doença/genética , Animais , Mapeamento Cromossômico , Genótipo , Humanos , Modelos Biológicos , Fenótipo
19.
J Hypertens ; 37(5): 985-996, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30308595

RESUMO

BACKGROUND: Combined congenic breeding and microarray gene expression profiling previously identified glutathione S-transferase µ-type 1 (Gstm1) as a positional and functional candidate gene for blood pressure (BP) regulation in the stroke-prone spontaneously hypertensive (SHRSP) rat. Renal Gstm1 expression in SHRSP rats is significantly reduced when compared with normotensive Wistar Kyoto (WKY) rats. As Gstm1 plays an important role in the secondary defence against oxidative stress, significantly lower expression levels may be functionally relevant in the development of hypertension. The aim of this study was to investigate the role of Gstm1 in BP regulation and oxidative stress by transgenic overexpression of the Gstm1 gene. METHOD: Two independent Gstm1 transgenic SHRSP lines were generated by microinjecting SHRSP embryos with a linear construct controlled by the EF-1α promoter encoding WKY Gstm1 cDNA [SHRSP-Tg(Gstm1)1 and SHRSP-Tg(Gstm1)2]. RESULTS: Transgenic rats exhibit significantly reduced BP and pulse pressure when compared with SHRSP [systolic: SHRSP 205.2 ±â€Š3.7 mmHg vs. SHRSP-Tg(Gstm1)1 175.5 ±â€Š1.6 mmHg and SHRSP-Tg(Gstm1)2 172 ±â€Š3.2 mmHg, P < 0.001; pulse pressure: SHRSP 58.4 ±â€Š0.73 mmHg vs. SHRSP-Tg(Gstm1)1 52.7 ±â€Š0.19 mmHg and SHRSP-Tg(Gstm1)2 40.7 ±â€Š0.53 mmHg, P < 0.001]. Total renal and aortic Gstm1 expression in transgenic animals was significantly increased compared with SHRSP [renal relative quantification (RQ): SHRSP-Tg(Gstm1)1 1.95 vs. SHRSP 1.0, P < 0.01; aorta RQ: SHRSP-Tg(Gstm1)1 2.8 vs. SHRSP 1.0, P < 0.05]. Renal lipid peroxidation (malondialdehyde: protein) and oxidized : reduced glutathione ratio levels were significantly reduced in both transgenic lines when compared with SHRSP [malondialdehyde: SHRSP 0.04 ±â€Š0.009 µmol/l vs. SHRSP-Tg(Gstm1)1 0.024 ±â€Š0.002 µmol/l and SHRSP-Tg(Gstm1)2 0.021 ±â€Š0.002 µmol/l; (oxidized : reduced glutathione ratio): SHRSP 5.19 ±â€Š2.26 µmol/l vs. SHRSP-Tg(Gstm1)1 0.17 ±â€Š0.11 µmol/l and SHRSP-Tg(Gstm1)2 0.47 ±â€Š0.22 µmol/l]. Transgenic SHRSP rats containing the WKY Gstm1 gene demonstrate significantly lower BP, reduced oxidative stress and improved levels of renal Gstm1 expression. CONCLUSION: These data support the hypothesis that reduced renal Gstm1 plays a role in the development of hypertension.


Assuntos
Pressão Sanguínea/genética , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Hipertensão/genética , Estresse Oxidativo/genética , Animais , Animais Geneticamente Modificados , Aorta/metabolismo , Glutationa/metabolismo , Hipertensão/fisiopatologia , Rim/metabolismo , Peroxidação de Lipídeos , Masculino , Malondialdeído/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Transgênicos , Sístole
20.
J Hypertens ; 26(7): 1343-52, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18551009

RESUMO

OBJECTIVES: Glutathione S-transferases are involved in defences against oxidative stress. We have recently demonstrated reduced expression of glutathione S-transferase mu type 1 (Gstm1) in a rat model of hypertension. Here, we examine the association between GSTM variants and hypertension in human. METHODS: We screened 83 patients with hypertension and 46 controls for single nucleotide polymorphisms in GSTM genes by TaqMan single nucleotide polymorphism genotyping assays and DNA sequencing. We then genotyped 753 trios from the Medical Research Council British Genetics of Hypertension Study transmission disequilibrium test cohort for 10 single nucleotide polymorphisms and the GSTM1 deletion and examined renal GSTM expression in a cohort of 27 hypertensive and 18 normotensive subjects. Finally, we attempted to replicate our findings in 1675 cases and 1654 controls from the Medical Research Council British Genetics of Hypertension Study case-control cohort. RESULTS: We identified two major linkage disequilibrium blocks including GSTM4/GSTM2 and GSTM5/GSTM3 separated by the GSTM1 gene. In the British Genetics of Hypertension transmission disequilibrium test resource, a single nucleotide polymorphism in the 3' region of GSTM5 (rs11807) was found to be associated with hypertension (P = 0.01) with the T-allele being over-transmitted to hypertensive offspring. GSTM5 mRNA expression was found to be reduced in kidney tissue of subjects homozygous for the T-allele of rs11807 as compared to C-allele homozygous and CT heterozygous subjects (P = 0.02). Nevertheless, rs11807 was not associated with hypertension in the British Genetics of Hypertension case-control cohort (P = 0.61). CONCLUSION: Our studies do not provide an evidence of an association of GSTM gene variants with hypertension in humans. They, however, illustrate the essential role of replication of initial results in a second cohort.


Assuntos
Glutationa Transferase/genética , Hipertensão/genética , Adulto , Feminino , Genótipo , Humanos , Desequilíbrio de Ligação , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA