Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37446003

RESUMO

Vascular access is the lifeline for patients receiving haemodialysis as kidney replacement therapy. As a surgically created arteriovenous fistula (AVF) provides a high-flow conduit suitable for cannulation, it remains the vascular access of choice. In order to use an AVF successfully, the luminal diameter and the vessel wall of the venous outflow tract have to increase. This process is referred to as AVF maturation. AVF non-maturation is an important limitation of AVFs that contributes to their poor primary patency rates. To date, there is no clear overview of the overall role of the extracellular matrix (ECM) in AVF maturation. The ECM is essential for vascular functioning, as it provides structural and mechanical strength and communicates with vascular cells to regulate their differentiation and proliferation. Thus, the ECM is involved in multiple processes that regulate AVF maturation, and it is essential to study its anatomy and vascular response to AVF surgery to define therapeutic targets to improve AVF maturation. In this review, we discuss the composition of both the arterial and venous ECM and its incorporation in the three vessel layers: the tunica intima, media, and adventitia. Furthermore, we examine the effect of chronic kidney failure on the vasculature, the timing of ECM remodelling post-AVF surgery, and current ECM interventions to improve AVF maturation. Lastly, the suitability of ECM interventions as a therapeutic target for AVF maturation will be discussed.


Assuntos
Fístula Arteriovenosa , Derivação Arteriovenosa Cirúrgica , Falência Renal Crônica , Humanos , Falência Renal Crônica/terapia , Diálise Renal , Matriz Extracelular
2.
Nat Rev Nephrol ; 17(12): 814-826, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34381199

RESUMO

Circular RNAs (circRNAs) are a class of endogenously expressed regulatory RNAs with a single-stranded circular structure. They are generated by back splicing and their expression can be tightly regulated by RNA binding proteins. Cytoplasmic circRNAs can function as molecular sponges that inhibit microRNA-target interactions and protein function or as templates for the efficient generation of peptides via rolling circle amplification. They can also act as molecular scaffolds that enhance the reaction kinetics of enzyme-substrate interactions. In the nucleus, circRNAs might facilitate chromatin modifications and promote gene expression. CircRNAs are resistant to degradation and can be packaged in extracellular vesicles and transported in the circulation. Initial studies suggest that circRNAs have roles in kidney disease and associated cardiovascular complications. They have been implicated in hypertensive nephropathy, diabetic kidney disease, glomerular disease, acute kidney injury and kidney allograft rejection, as well as in microvascular and macrovascular complications of chronic kidney disease, including atherosclerotic vascular disease. In addition, several circRNAs have been reported to have oncogenic or tumour suppressor roles or to regulate drug resistance in kidney cancer. The available data suggest that circRNAs could be promising diagnostic and/or prognostic biomarkers and potential therapeutic targets for kidney disease, cardiovascular disease and kidney cancer.


Assuntos
Doenças Cardiovasculares , Neoplasias Renais , MicroRNAs , Humanos , MicroRNAs/genética , RNA Circular , Proteínas de Ligação a RNA
3.
Eur J Endocrinol ; 185(4): 539-552, 2021 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-34342596

RESUMO

OBJECTIVE: Sex steroid hormones like estrogens have a key role in the regulation of energy homeostasis and metabolism. In transwomen, gender-affirming hormone therapy like estradiol (in combination with antiandrogenic compounds) could affect metabolism as well. Given that the underlying pathophysiological mechanisms are not fully understood, this study assessed circulating estradiol-driven microRNAs (miRs) in transwomen and their regulation of genes involved in metabolism in mice. METHODS: Following plasma miR-sequencing (seq) in a transwomen discovery (n = 20) and validation cohort (n = 30), we identified miR-224 and miR-452. Subsequent systemic silencing of these miRs in male C57Bl/6 J mice (n = 10) was followed by RNA-seq-based gene expression analysis of brown and white adipose tissue in conjunction with mechanistic studies in cultured adipocytes. RESULTS: Estradiol in transwomen lowered plasma miR-224 and -452 carried in extracellular vesicles (EVs) while their systemic silencing in mice and cultured adipocytes increased lipogenesis (white adipose) but reduced glucose uptake and mitochondrial respiration (brown adipose). In white and brown adipose tissue, differentially expressed (miR target) genes are associated with lipogenesis (white adipose) and mitochondrial respiration and glucose uptake (brown adipose). CONCLUSION: This study identified an estradiol-drive post-transcriptional network that could potentially offer a mechanistic understanding of metabolism following gender-affirming estradiol therapy.


Assuntos
Micropartículas Derivadas de Células/genética , Estradiol/fisiologia , MicroRNAs/genética , Transexualidade , Adipócitos/efeitos dos fármacos , Adipócitos/fisiologia , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Adulto , Animais , Micropartículas Derivadas de Células/efeitos dos fármacos , Micropartículas Derivadas de Células/metabolismo , Estudos de Coortes , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Estradiol/sangue , Estradiol/farmacologia , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Terapia de Reposição Hormonal , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Interferência de RNA/efeitos dos fármacos , Pessoas Transgênero , Transexualidade/genética , Transexualidade/metabolismo , Adulto Jovem
4.
Sci Rep ; 10(1): 13945, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32811874

RESUMO

Left ventricular diastolic dysfunction (LVDD) and heart failure with preserved ejection fraction (HFpEF) are microcirculation defects following diabetes mellitus (DM). Unrecognized HFpEF is more prevalent in women with diabetes compared to men with diabetes and therefore sex-specific diagnostic strategies are needed. Previously, we demonstrated altered plasma miRs in DM patients with microvascular injury [defined by elevated plasma Angiopoietin-2 (Ang-2) levels]. This study hypothesized the presence of sex-differences in plasma miRs and Ang-2 in diabetic (female) patients with LVDD or HFpEF. After a pilot study, we assessed 16 plasma miRs in patients with LVDD (n = 122), controls (n = 244) and female diabetic patients (n = 10). Subsequently, among these miRs we selected and measured plasma miR-34a, -224 and -452 in diabetic HFpEF patients (n = 53) and controls (n = 52). In LVDD patients, miR-34a associated with Ang-2 levels (R2 0.04, R = 0.21, p = 0.001, 95% CI 0.103-0.312), with plasma levels being diminished in patients with DM, while women with an eGFR < 60 ml/min and LVDD had lower levels of miR-34a, -224 and -452 compared to women without an eGFR < 60 ml/min without LVDD. In diabetic HFpEF women (n = 28), plasma Ang-2 levels and the X-chromosome located miR-224/452 cluster increased compared to men. We conclude that plasma miR-34a, -224 and -452 display an association with the microvascular injury marker Ang-2 and are particularly targeted to women with LVDD or HFpEF.


Assuntos
Insuficiência Cardíaca/genética , MicroRNAs/genética , Disfunção Ventricular Esquerda/genética , Adulto , Idoso , Angiopoietina-2/análise , Angiopoietina-2/sangue , Biomarcadores/sangue , Complicações do Diabetes/genética , Diabetes Mellitus/genética , Feminino , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Caracteres Sexuais , Volume Sistólico/genética , Volume Sistólico/fisiologia , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda/genética , Função Ventricular Esquerda/fisiologia
5.
Am J Transplant ; 20(12): 3451-3461, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32353171

RESUMO

Simultaneous pancreas-kidney transplantation (SPKT) replaces kidney function and restores endogenous insulin secretion in patients with diabetic nephropathy (DN). Here, we aimed to identify circulating long noncoding RNAs (lncRNAs) that are associated with DN and vascular injury in the context of SPKT. Based on a pilot study and a literature-based selection of vascular injury-related lncRNAs, we assessed 9 candidate lncRNAs in plasma samples of patients with diabetes mellitus with a kidney function >35 mL/min/1.73 m2 (DM; n = 12), DN (n = 14), SPKT (n = 35), healthy controls (n = 15), and renal transplant recipients (KTx; n = 13). DN patients were also studied longitudinally before and 1, 6, and 12 months after SPKT. Of 9 selected lncRNAs, we found MALAT1, LIPCAR, and LNC-EPHA6 to be higher in DN compared with healthy controls. SPKT caused MALAT1, LIPCAR, and LNC-EPHA6 to normalize to levels of healthy controls, which was confirmed in the longitudinal study. In addition, we observed a strong association between MALAT1, LNC-EPHA6, and LIPCAR and vascular injury marker soluble thrombomodulin and a subset of angiogenic microRNAs (miR-27a, miR-130b, miR-152, and miR-340). We conclude that specific circulating lncRNAs associate with DN-related vascular injury and normalize after SPKT, suggesting that lncRNAs may provide a promising novel monitoring strategy for vascular integrity in the context of SPKT.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Transplante de Rim , MicroRNAs , Transplante de Pâncreas , RNA Longo não Codificante , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/cirurgia , Humanos , Transplante de Rim/efeitos adversos , Estudos Longitudinais , Masculino , Pâncreas , Projetos Piloto , RNA Longo não Codificante/genética
6.
Clin Sci (Lond) ; 133(2): 239-252, 2019 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-30617188

RESUMO

Kidney fibrosis is the common pathophysiological mechanism in end-stage renal disease characterized by excessive accumulation of myofibroblast-derived extracellular matrix. Natriuretic peptides have been demonstrated to have cyclic guanosine monophosphate (cGMP)-dependent anti-fibrotic properties likely due to interference with pro-fibrotic tissue growth factor ß (TGF-ß) signaling. However, in vivo, natriuretic peptides are rapidly degraded by neutral endopeptidases (NEP). In a unilateral ureteral obstruction (UUO) mouse model for kidney fibrosis we assessed the anti-fibrotic effects of SOL1, an orally active compound that inhibits NEP and endothelin-converting enzyme (ECE). Mice (n=10 per group) subjected to UUO were treated for 1 week with either solvent, NEP-/ECE-inhibitor SOL1 (two doses), reference NEP-inhibitor candoxatril or the angiotensin II receptor type 1 (AT1)-antagonist losartan. While NEP-inhibitors had no significant effect on blood pressure, they did increase urinary cGMP levels as well as endothelin-1 (ET-1) levels. Immunohistochemical staining revealed a marked decrease in renal collagen (∼55% reduction, P<0.05) and α-smooth muscle actin (α-SMA; ∼40% reduction, P<0.05). Moreover, the number of α-SMA positive cells in the kidneys of SOL1-treated groups inversely correlated with cGMP levels consistent with a NEP-dependent anti-fibrotic effect. To dissect the molecular mechanisms associated with the anti-fibrotic effects of NEP inhibition, we performed a 'deep serial analysis of gene expression (Deep SAGE)' transcriptome and targeted metabolomics analysis of total kidneys of all treatment groups. Pathway analyses linked increased cGMP and ET-1 levels with decreased nuclear receptor signaling (peroxisome proliferator-activated receptor [PPAR] and liver X receptor/retinoid X receptor [LXR/RXR] signaling) and actin cytoskeleton organization. Taken together, although our transcriptome and metabolome data indicate metabolic dysregulation, our data support the therapeutic potential of NEP inhibition in the treatment of kidney fibrosis via cGMP elevation and reduced myofibroblast formation.


Assuntos
Benzazepinas/farmacologia , Nefropatias/prevenção & controle , Rim/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Neprilisina/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Obstrução Ureteral/tratamento farmacológico , Animais , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Rim/enzimologia , Rim/patologia , Nefropatias/enzimologia , Nefropatias/genética , Nefropatias/patologia , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Células NIH 3T3 , Neprilisina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Obstrução Ureteral/enzimologia , Obstrução Ureteral/genética , Obstrução Ureteral/patologia
7.
Cardiovasc Res ; 114(2): 210-225, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29186452

RESUMO

Cardiovascular disease (CVD) is the primary cause of death among men and women worldwide. Nevertheless, our comprehension of how CVD progresses in women and elicits clinical outcomes is lacking, leading CVD to be under-diagnosed and under-treated in women. A clear example of this differential presentation of CVD pathophysiologies in females is the strikingly higher prevalence of heart failure with preserved ejection fraction (HFpEF). Women with a history of pre-eclampsia or those who present with co-morbidities such as obesity, hypertension, and diabetes mellitus are at increased risk of developing HFpEF. Long understood to be a critical CVD risk factor, our understanding of how gender differentially affects the development of CVD has been greatly expanded by extensive genomic and transcriptomic studies. These studies uncovered a pivotal role for differential microRNA (miRNA) expression in response to systemic inflammation, where their co-ordinated expression forms a post-transcriptional regulatory network that instigates microcirculation defects. Importantly, the potential sex-biased expression of the given miRNAs may explain sex-specific cardiovascular pathophysiologies in women, such as HFpEF. Sex-biased miRNAs are regulated by oestrogen (E2) in their transcription and processing or are expressed from loci on the X-chromosome due to incomplete X-chromosome inactivation. Interestingly, while E2-induced miRNAs predominantly appear to serve protective functions, it could be argued that many X-linked miRNAs have been found to challenge microvascular and myocardial integrity. Therefore, menopausal E2 deficiency, resulting in protective miRNA loss, and the augmentation of X-linked miRNA expression, may well contribute to the molecular mechanisms that underlie the female-specific cardiovascular aetiology in HFpEF.


Assuntos
Disparidades nos Níveis de Saúde , Insuficiência Cardíaca/genética , MicroRNAs/genética , Volume Sistólico , Função Ventricular Esquerda , Fatores Etários , Animais , Cromossomos Humanos X , Comorbidade , Estradiol/metabolismo , Feminino , Regulação da Expressão Gênica , Insuficiência Cardíaca/epidemiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Menopausa/genética , Menopausa/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Prevalência , Medição de Risco , Fatores de Risco , Fatores Sexuais , Remodelação Ventricular , Inativação do Cromossomo X
8.
PLoS One ; 12(12): e0187118, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29206835

RESUMO

We recently demonstrated that the adult human kidney cortex contains a perivascular stromal cell (kPSC) that shows organotypic properties and is important for repair and stabilisation of kidney function. Not only the kidney cortex but also the kidney capsule contains stromal cells that are important for the three dimensional organisation of the kidney during nephrogenesis. They provide the barrier function of the capsule which is critical for homeostatic processes such as pressure natriuresis. We postulated that stromal cells derived from the kidney capsule may therefore also have specific properties and functions. To this end, we isolated these capsule mesenchymal stromal cells (cMSC) from human cadaveric kidneys that were not suitable for transplantation. There were several similarities between cMSCs and kPSCs including support of vascular plexus formation, phenotypic marker expression and resistance against myofibroblast transformation. However, compared to kPSCs, cMSCs showed distinct mRNA and miRNA expression profiles, showed increased immunosuppressive capacity, and displayed strongly reduced HGF production, contributing to the inability to enhance kidney epithelial repair. Therefore cMSCs are a distinct, novel human kidney-derived MSC-population and these data underpin the large functional diversity of phenotypic similar stromal cells in relation to their anatomic site, even within one organ.


Assuntos
Rim/citologia , Células-Tronco Mesenquimais/citologia , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Técnicas de Cocultura , Perfilação da Expressão Gênica , Humanos , Rim/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , RNA Mensageiro/genética , Fator de Crescimento Transformador beta/metabolismo
9.
Kidney Int ; 89(6): 1268-80, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27165825

RESUMO

Chronic kidney disease is associated with progressive renal fibrosis, where perivascular cells give rise to the majority of α-smooth muscle actin (α-SMA) positive myofibroblasts. Here we sought to identify pericytic miRNAs that could serve as a target to decrease myofibroblast formation. Kidney fibrosis was induced in FoxD1-GC;Z/Red-mice by unilateral ureteral obstruction followed by FACS sorting of dsRed-positive FoxD1-derivative cells and miRNA profiling. MiR-132 selectively increased 21-fold during pericyte-to-myofibroblast formation, whereas miR-132 was only 2.5-fold up in total kidney lysates (both in obstructive and ischemia-reperfusion injury). MiR-132 silencing during obstruction decreased collagen deposition (35%) and tubular apoptosis. Immunohistochemistry, Western blot, and qRT-PCR confirmed a similar decrease in interstitial α-SMA(+) cells. Pathway analysis identified a rate-limiting role for miR-132 in myofibroblast proliferation that was confirmed in vitro. Indeed, antagomir-132-treated mice displayed a reduction in the number of proliferating Ki67(+) interstitial myofibroblasts. Interestingly, this was selective for the interstitial compartment and did not impair the reparative proliferation of tubular epithelial cells, as evidenced by an increase in Ki67(+) epithelial cells, as well as increased phospho-RB1, Cyclin-A and decreased RASA1, p21 levels in kidney lysates. Additional pathway and gene expression analyses suggest miR-132 coordinately regulates genes involved in TGF-ß signaling (Smad2/Smad3), STAT3/ERK pathways, and cell proliferation (Foxo3/p300). Thus, silencing miR-132 counteracts the progression of renal fibrosis by selectively decreasing myofibroblast proliferation and could potentially serve as a novel antifibrotic therapy.


Assuntos
Proliferação de Células/genética , Rim/patologia , MicroRNAs/genética , Miofibroblastos/fisiologia , Insuficiência Renal Crônica/patologia , Actinas/metabolismo , Animais , Antagomirs/genética , Apoptose , Linhagem Celular , Colágeno/metabolismo , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Fibrose , Humanos , Imuno-Histoquímica , Túbulos Renais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Pericitos/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fator de Crescimento Transformador beta
10.
Sci Rep ; 6: 21643, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26905650

RESUMO

Proper regulation of endothelial cell-cell contacts is essential for physiological functioning of the endothelium. Interendothelial junctions are actively involved in the control of vascular leakage, leukocyte diapedesis, and the initiation and progression of angiogenesis. We found that the RNA-binding protein quaking is highly expressed by endothelial cells, and that its expression was augmented by prolonged culture under laminar flow and the transcription factor KLF2 binding to the promoter. Moreover, we demonstrated that quaking directly binds to the mRNA of VE-cadherin and ß-catenin and can induce mRNA translation mediated by the 3'UTR of these genes. Reduced quaking levels attenuated VE-cadherin and ß-catenin expression and endothelial barrier function in vitro and resulted in increased bradykinin-induced vascular leakage in vivo. Taken together, we report that quaking is essential in maintaining endothelial barrier function. Our results provide novel insight into the importance of post-transcriptional regulation in controlling vascular integrity.


Assuntos
Antígenos CD/genética , Caderinas/genética , Células Endoteliais da Veia Umbilical Humana/fisiologia , Proteínas de Ligação a RNA/fisiologia , beta Catenina/genética , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar , Feminino , Expressão Gênica , Células HEK293 , Humanos , Fatores de Transcrição Kruppel-Like/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ativação Transcricional , beta Catenina/metabolismo
11.
Diabetologia ; 59(4): 853-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26768002

RESUMO

AIMS/HYPOTHESIS: High levels of circulating mannan-binding lectin (MBL) are associated with the development of diabetic nephropathy and hyperglycaemia-induced vasculopathy. Here, we aimed to assess the effect of glycaemic control on circulating levels of MBL and the relationship of these levels with vascular damage. METHODS: We assessed MBL levels and corresponding MBL2 genotype, together with vascular endothelial growth factor (VEGF) levels as a marker of vascular damage, in type 1 diabetes patients with diabetic nephropathy before and after simultaneous pancreas-kidney (SPK) transplantation. We included diabetic nephropathy patients (n = 21), SPK patients (n = 37), healthy controls (n = 19), type 1 diabetes patients (n = 15) and diabetic nephropathy patients receiving only a kidney transplant (n = 15). Fourteen diabetic nephropathy patients were followed up for 12 months after SPK. RESULTS: We found elevated circulating MBL levels in diabetic nephropathy patients, and a trend towards elevated circulating MBL levels in type 1 diabetes patients, compared with healthy control individuals. MBL levels in SPK patients completely normalised and our data indicate that this predominantly occurs in patients with a polymorphism in the MBL2 gene. By contrast, MBL levels in kidney transplant only patients remained elevated, suggesting that glycaemic control but not reversal of renal failure is associated with decreased MBL levels. In line, levels of glucose and HbA1c, but not creatinine levels and estimated GFR, were correlated with MBL levels. VEGF levels were associated with levels of MBL and HbA1c in an MBL-polymorphism-dependent manner. CONCLUSIONS/INTERPRETATION: Taken together, circulating MBL levels are associated with diabetic nephropathy and are dependent on glycaemic control, possibly in an MBL2-genotype-dependent manner.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/cirurgia , Transplante de Rim , Lectina de Ligação a Manose/sangue , Transplante de Pâncreas , Fator A de Crescimento do Endotélio Vascular/sangue , Estudos Transversais , Diabetes Mellitus Tipo 1/sangue , Feminino , Genótipo , Humanos , Masculino , Lectina de Ligação a Manose/genética , Fator A de Crescimento do Endotélio Vascular/genética
12.
Curr Vasc Pharmacol ; 13(3): 341-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-23713864

RESUMO

MicroRNAs are negative regulators of gene expression that have been shown to be essential elements in the coordination of complex regulatory pathways. One of these short non-coding RNAs, microRNA-126, is highly enriched in the vascular endothelium and was shown to play distinct roles in angiogenesis, vasculogenesis and endothelial inflammation. Abrogation of this microRNA leads to severe complications in the response in vascular development as well as vital repair mechanisms carried out by endothelial cells. Interestingly, recent data suggest that the homeostatic role of microRNA-126 may reach far beyond its endothelial functions as this microRNA was also found to be present in cells of the hematopoietic system and in microvesicles or 'free-form' in the periphery. MicroRNA-126 is controlling the fate and/or function of a variety of cells differentiating from the hematopoietic lineage, including megakaryocytes and erythrocytes. Recent studies identified circulating microRNA-126 as a biomarker for myocardial injury and vascular damage in diabetes. Furthermore, reports have suggested a protective role of circulating microRNA-126 in murine models of organ ischemia. Here, we review current insights in the role of microRNA-126 in vascular homeostasis and conclude that this microRNA may serve to integrate and facilitate both local as well as systemic functions in vascular maintenance and repair.


Assuntos
Vasos Sanguíneos/fisiologia , Homeostase/genética , Homeostase/fisiologia , MicroRNAs/fisiologia , Animais , Humanos , Camundongos , MicroRNAs/genética , Doenças Vasculares/genética , Doenças Vasculares/patologia
13.
Microrna ; 3(3): 144-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25541911

RESUMO

Integrity of the capillary network in the kidney is essential in the recovery from ischemia/ reperfusion injury (IRI), a phenomenon central to kidney transplantation and acute kidney injury. MicroRNA- 126 (miR-126) is known to be important in maintaining vascular homeostasis by facilitating vascular regeneration and modulating the mobilization of vascular progenitor cells. Stromal cell-derived factor 1 (SDF-1), important in the mobilization of vascular progenitor cells, is a direct target of miR-126 and modulation of miR-126 was previously shown to affect the number of circulating Sca-1(+)/Lin(-) vascular progenitor cells in a mouse model for hind limb ischemia. Here, we assessed the in vivo contribution of miR-126 to progenitor cell mobilization and kidney function following IRI in mice. A three day follow up of blood urea levels following kidney IRI demonstrated that systemic antagomir silencing of miR-126 did not impact the loss or subsequent restoration of kidney function. However, whole kidney lysates displayed elevated gene expression levels of Sdf-1, Vegf-A and eNOS after IRI as a result of systemic silencing of miR-126. Furthermore, FACS-analysis on whole blood three days after surgery revealed a marked up regulation of the number of circulating Sca-1(+)/Lin(-) progenitor cells in the antagomir-126 treated mice, in an ischemia dependent manner. Our data indicate that silencing of miR-126 can enhance renal expression of Sdf-1 after IRI, leading to the mobilization of vascular progenitor cells into the circulation.


Assuntos
Antígenos Ly/metabolismo , Quimiocina CXCL12/metabolismo , Células Progenitoras Endoteliais/metabolismo , Rim/metabolismo , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Antígenos Ly/genética , Quimiocina CXCL12/genética , Células Progenitoras Endoteliais/citologia , Inativação Gênica , Rim/irrigação sanguínea , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
J Am Soc Nephrol ; 25(8): 1710-22, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24610930

RESUMO

Ischemia/reperfusion injury (IRI) is a central phenomenon in kidney transplantation and AKI. Integrity of the renal peritubular capillary network is an important limiting factor in the recovery from IRI. MicroRNA-126 (miR-126) facilitates vascular regeneration by functioning as an angiomiR and by modulating mobilization of hematopoietic stem/progenitor cells. We hypothesized that overexpression of miR-126 in the hematopoietic compartment could protect the kidney against IRI via preservation of microvascular integrity. Here, we demonstrate that hematopoietic overexpression of miR-126 increases neovascularization of subcutaneously implanted Matrigel plugs in mice. After renal IRI, mice overexpressing miR-126 displayed a marked decrease in urea levels, weight loss, fibrotic markers, and injury markers (such as kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin). This protective effect was associated with a higher density of the peritubular capillary network in the corticomedullary junction and increased numbers of bone marrow-derived endothelial cells. Hematopoietic overexpression of miR-126 increased the number of circulating Lin(-)/Sca-1(+)/cKit(+) hematopoietic stem and progenitor cells. Additionally, miR-126 overexpression attenuated expression of the chemokine receptor CXCR4 on Lin(-)/Sca-1(+)/cKit(+) cells in the bone marrow and increased renal expression of its ligand stromal cell-derived factor 1, thus favoring mobilization of Lin(-)/Sca-1(+)/cKit(+) cells toward the kidney. Taken together, these results suggest overexpression of miR-126 in the hematopoietic compartment is associated with stromal cell-derived factor 1/CXCR4-dependent vasculogenic progenitor cell mobilization and promotes vascular integrity and supports recovery of the kidney after IRI.


Assuntos
Injúria Renal Aguda/prevenção & controle , Células-Tronco Hematopoéticas/fisiologia , Rim/irrigação sanguínea , MicroRNAs/fisiologia , Neovascularização Fisiológica/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Movimento Celular/fisiologia , Quimiocina CXCL12/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Receptores CXCR4/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
15.
Cardiovasc Res ; 92(3): 449-55, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21856785

RESUMO

AIMS: MicroRNA-126 (miR-126), which is enriched in endothelial cells, plays a role in angiogenesis. Based on the seed sequence, miR-126 can also be predicted to regulate vasculogenesis by modulating the endothelial expression of stromal cell-derived factor-1 (SDF-1). METHODS AND RESULTS: Using miR-reporter constructs, we first validated that miR-126 inhibits SDF-1 expression in endothelial cells in vitro. Next, we investigated the potential relevance of this observation with respect to the mobilization of progenitor cells. For this, we studied the migration of human CD34+ progenitor cells towards chemotactic factors present in endothelial cell-conditioned medium. Antagomir-induced silencing of miR-126 elevated SDF-1 expression by human umbilical vein endothelial cells and enhanced migration of the CD34+ cells. In a murine model of hind limb ischaemia, a striking increase in the number of circulating Sca-1(+)/Lin(-) progenitor cells in antagomir-126-treated mice was observed when compared with scramblemir-treated controls. Immunohistochemical staining of capillaries in the post-ischaemic gastrocnemius muscle of miR-126-silenced mice revealed elevated SDF-1 expressing CD31-positive capillaries, whereas a mobilizing effect of miR-126 inhibition was not detected in healthy control animals. CONCLUSION: miR-126 can regulate the expression of SDF-1 in endothelial cells. In the context of an ischaemic event, systemic silencing of miR-126 leads to the mobilization of Sca-1(+)/Lin(-) progenitor cells into the peripheral circulation, potentially in response to elevated SDF-1 expression by endothelial cells present in the ischaemic tissue.


Assuntos
Antígenos Ly/metabolismo , Quimiocina CXCL12/metabolismo , Quimiotaxia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Isquemia/metabolismo , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Músculo Esquelético/irrigação sanguínea , Células-Tronco/metabolismo , Regiões 3' não Traduzidas , Animais , Antígenos CD34/metabolismo , Células Cultivadas , Quimiocina CXCL12/genética , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Genes Reporter , Membro Posterior , Humanos , Imuno-Histoquímica , Isquemia/genética , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Oligonucleotídeos/administração & dosagem , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA