Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Int J Obes (Lond) ; 46(6): 1222-1233, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35256761

RESUMO

INTRODUCTION: Obesity is a health burden that impairs cellular processes. Mesenchymal stem/stromal cells (MSCs) are endowed with reparative properties and can ameliorate renal injury. Obesity impairs human MSC function in-vitro, but its effect on their in-vivo reparative potency remains unknown. SUBJECTS AND METHODS: Abdominal adipose tissue-derived MSC were harvested from patients without ('lean') or with obesity ('obese') (body mass index <30 or ≥30 kg/m2, respectively) during kidney donation or bariatric surgery, respectively. MSC (5 × 105/200 µL) or vehicle were then injected into 129S1 mice 2 weeks after renal artery stenosis (RAS) or sham surgery (n = 8/group). Two weeks later, mice underwent magnetic resonance imaging to assess renal perfusion and oxygenation in-vivo, and kidneys then harvested for ex-vivo studies. RESULTS: Similar numbers of lean and obese-MSCs engrafted in stenotic mouse kidneys. Vehicle-treated RAS mice had reduced stenotic-kidney cortical and medullary perfusion and oxygenation. Lean (but not obese) MSC normalized ischemic kidney cortical perfusion, whereas both effectively mitigated renal hypoxia. Serum creatinine and blood pressure were elevated in RAS mice and lowered only by lean-MSC. Both types of MSCs alleviated stenotic-kidney fibrosis, but lean-MSC more effectively than obese-MSC. MSC senescence-associated beta-gal activity, and gene expression of p16, p21, and vascular endothelial growth factor correlated with recipient kidney perfusion and tissue injury, linking MSC characteristics with their in-vivo reparative capacity. DISCUSSION: Human obesity impairs the reparative properties of adipose-tissue-derived MSCs, possibly by inducing cellular senescence. Dysfunction and senescence of the endogenous MSC repair system in patients with obesity may warrant targeting interventions to restore MSC vitality.


Assuntos
Células-Tronco Mesenquimais , Obstrução da Artéria Renal , Animais , Humanos , Rim/patologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Obesidade/metabolismo , Obstrução da Artéria Renal/metabolismo , Obstrução da Artéria Renal/patologia , Fator A de Crescimento do Endotélio Vascular
2.
J Tissue Eng Regen Med ; 16(6): 550-558, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35319825

RESUMO

Chronic ischemia triggers senescence in renal tubules and at least partly mediates kidney dysfunction and damage through a p16Ink4a -related mechanism. We previously showed that mesenchymal stromal/stem cells (MSCs) delivered systemically do not effectively decrease cellular senescence in stenotic murine kidneys. We hypothesized that selective MSC targeting to injured kidneys using an anti-KIM1 antibody (KIM-MSC) coating would enhance their ability to abrogate cellular senescence in murine renal artery stenosis (RAS). KIM-MSC were injected into transgenic INK-ATTAC mice, which are amenable for selective eradication of p16Ink4a+ cells, 4 weeks after induction of unilateral RAS. To determine whether KIM-MSC abolish p16Ink4a -dependent cellular senescence, selective clearance of p16Ink4a+ cells was induced in a subgroup of RAS mice using AP20187 over 3 weeks prior to KIM-MSC injection. Two weeks after KIM-MSC aortic injection, renal senescence, function, and tissue damage were assessed. KIM-MSC delivery decreased gene expression of senescence and senescence-associated secretory phenotype factors, and improved micro-MRI-derived stenotic-kidney glomerular filtration rate and perfusion. Renal fibrosis and tubular injury also improved after KIM-MSC treatment. Yet, their efficacy was slightly augmented by prior elimination of p16Ink4a+ senescent cells. Therefore, selective targeting of MSC to the injured kidney markedly improves their senolytic potency in murine RAS, despite incomplete eradication of p16+ cells. KIM-MSC may constitute a useful therapeutic strategy in chronic renal ischemic injury.


Assuntos
Células-Tronco Mesenquimais , Obstrução da Artéria Renal , Animais , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Isquemia/metabolismo , Rim/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Obstrução da Artéria Renal/metabolismo
3.
Am J Physiol Renal Physiol ; 320(3): F454-F463, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33554782

RESUMO

Tumor necrosis factor (TNF)-α-induced gene/protein (TSG)-6 regulates the immunomodulatory properties of mesenchymal stem cells (MSCs), but its ability to protect the ischemic kidney is unknown. In a swine model of renal artery stenosis (RAS) and metabolic syndrome (MetS), we assessed the contribution of TSG-6 produced by MSCs to their immunomodulatory properties. Pigs were studied after 16 wk of diet-induced MetS and unilateral RAS and were either untreated or treated 4 wk earlier with intrarenal autologous adipose tissue-derived MSCs (n = 6 each). Lean, MetS, and RAS sham animals served as controls. We studied renal function in vivo (using computed tomography) and kidney histopathology and macrophage phenotype ex vivo. In vitro, TSG-6 levels were also measured in conditioned media of human MSCs incubated with TNF-α and levels of the tubular injury marker lactate dehydrogenase in conditioned media after coculturing macrophages with injured human kidney 2 (HK-2) cells with or without TSG-6. The effects of TSG-6 on macrophage phenotype (M1/M2), adhesion, and migration were also determined. MetS + RAS showed increased M1 macrophages and renal vein TNF-α levels. After MSC delivery, renal vein TSG-6 increased and TNF-α decreased, the M1-to-M2 ratio decreased, renal function improved, and fibrosis was alleviated. In vitro, TNF-α increased TSG-6 secretion by human MSCs. TSG-6 decreased lactate dehydrogenase release from injured HK-2 cells, increased expression of macrophage M2 markers, and reduced M1 macrophage adhesion and migration. Therefore, TSG-6 released from MSCs may decrease renal tubular cell injury, which is associated with regulating macrophage function and phenotype. These observations suggest that TSG-6 is endowed with renoprotective properties.NEW & NOTEWORTHY Tumor necrosis factor-α-induced gene/protein (TSG)-6 regulates the immunomodulatory properties of MSCs, but its ability to protect the ischemic kidney is unknown. In pigs with renal artery stenosis, we show that MSC delivery increased renal vein TSG-6, decreased kidney inflammatory macrophages, and improved renal function. In vitro, TSG-6 decreased inflammatory macrophages and tubular cell injury. Therefore, TSG-6 released from MSCs may decrease renal tubular cell injury, which is associated with regulating macrophage function and phenotype.


Assuntos
Células Epiteliais/citologia , Macrófagos/citologia , Células-Tronco Mesenquimais/citologia , Fenótipo , Obstrução da Artéria Renal/patologia , Animais , Técnicas de Cocultura , Citocinas/metabolismo , Humanos , Rim/metabolismo , Rim/patologia , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/metabolismo , Substâncias Protetoras/farmacologia , Obstrução da Artéria Renal/metabolismo , Suínos , Fator de Necrose Tumoral alfa/metabolismo
4.
Hypertension ; 77(2): 507-518, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33390051

RESUMO

Scattered tubular-like cells (STCs), dedifferentiated renal tubular epithelial cells, contribute to renal self-healing, but severe injury might blunt their effectiveness. We hypothesized that ischemic renovascular disease (RVD) induces senescence in STC and impairs their reparative potency. CD24+/CD133+ STCs were isolated from swine kidneys after 16 weeks of RVD or healthy controls. To test their reparative capabilities in injured kidneys, control or RVD-STC (5×105) were prelabeled and injected into the aorta of 2 kidneys, 1-clip (2k,1c) mice 2 weeks after surgery. Murine renal function and oxygenation were studied in vivo 2 weeks after injection using micro-magnetic resonance imaging, and fibrosis, tubulointerstitial injury, capillary density, and expression of profibrotic and inflammatory genes ex vivo. STC isolated from swine RVD kidneys showed increased gene expression of senescence and senescence-associated secretory phenotype markers and positive SA-ß-gal staining. Delivery of normal pig STCs in 2k,1c mice improved murine renal perfusion, blood flow, and glomerular filtration rate, and downregulated profibrotic and inflammatory gene expression. These renoprotective effects were blunted using STC harvested from RVD kidneys, which also failed to attenuate hypoxia, fibrosis, tubular injury, and capillary loss in injured mouse 2k,1c kidneys. Hence, RVD may induce senescence in endogenous STC and impair their reparative capacity. These observations implicate cellular senescence in the pathophysiology of ischemic kidney disease and support senolytic therapy to permit self-healing of senescent kidneys.


Assuntos
Senescência Celular/fisiologia , Rim/patologia , Obstrução da Artéria Renal/patologia , Insuficiência Renal/patologia , Animais , Células Cultivadas , Feminino , Fibrose/metabolismo , Fibrose/patologia , Rim/metabolismo , Camundongos , Obstrução da Artéria Renal/metabolismo , Insuficiência Renal/metabolismo , Suínos
5.
J Vis Exp ; (164)2020 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-33165324

RESUMO

Renal artery stenosis is a common condition in patients with coronary or peripheral vascular disease where the renin angiotensin aldosterone system (RAAS) is overactivated. In this context, there is a narrowing of the renal arteries that stimulate an increase in the expression and release of renin, the rate-limiting protease in RAAS. The resulting rise in renin expression is a known driver of renovascular hypertension, frequently associated with kidney injury and end organ damage. Thus, there is a great interest in developing novel treatments for this condition. The molecular and cellular mechanism of renin control in renal artery stenosis is not fully understood and warrants further investigation. To induce renal artery stenosis in mice, a modified 2 kidney 1 clip (2K1C) Goldblatt mouse model was developed. The right kidney was stenosed in wild type mice and sham operated mice were used as control. After renal artery stenosis, we determined renin expression and kidney injury. Kidneys were harvested, and fresh cortices were used to determine protein and mRNA expression of renin. This animal model is reproducible and can be used to study pathophysiological responses, molecular and cellular pathways involved in renovascular hypertension and kidney injury.


Assuntos
Injúria Renal Aguda/diagnóstico , Modelos Animais de Doenças , Rim/cirurgia , Lipocalina-2/metabolismo , Obstrução da Artéria Renal/fisiopatologia , Artéria Renal/fisiopatologia , Renina/metabolismo , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Animais , Pressão Sanguínea , Feminino , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obstrução da Artéria Renal/complicações , Obstrução da Artéria Renal/metabolismo
6.
J Am Heart Assoc ; 9(7): e014072, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32200719

RESUMO

Background Renal artery stenosis is a common cause of renal ischemia, contributing to the development of chronic kidney disease. To investigate the role of local CD40 expression in renal artery stenosis, Goldblatt 2-kidney 1-clip surgery was performed on hypertensive Dahl salt-sensitive rats (S rats) and genetically modified S rats in which CD40 function is abolished (Cd40mutant). Methods and Results Four weeks following the 2-kidney 1-clip procedure, Cd40mutant rats demonstrated significantly reduced blood pressure and renal fibrosis in the ischemic kidneys compared with S rat controls. Similarly, disruption of Cd40 resulted in reduced 24-hour urinary protein excretion in Cd40mutant rats versus S rat controls (46.2±1.9 versus 118.4±5.3 mg/24 h; P<0.01), as well as protection from oxidative stress, as indicated by increased paraoxonase activity in Cd40mutant rats versus S rat controls (P<0.01). Ischemic kidneys from Cd40mutant rats demonstrated a significant decrease in gene expression of the profibrotic mediator, plasminogen activator inhibitor-1 (P<0.05), and the proinflammatory mediators, C-C motif chemokine ligand 19 (P<0.01), C-X-C Motif Chemokine Ligand 9 (P<0.01), and interleukin-6 receptor (P<0.001), compared with S rat ischemic kidneys, as assessed by quantitative PCR assay. Reciprocal renal transplantation documented that CD40 exclusively expressed in the kidney contributes to ischemia-induced renal fibrosis. Furthermore, human CD40-knockout proximal tubule epithelial cells suggested that suppression of CD40 signaling significantly inhibited expression of proinflammatory and -fibrotic genes. Conclusions Taken together, our data suggest that activation of CD40 induces a significant proinflammatory and -fibrotic response and represents an attractive therapeutic target for treatment of ischemic renal disease.


Assuntos
Antígenos CD40/metabolismo , Isquemia/metabolismo , Rim/irrigação sanguínea , Rim/metabolismo , Mutação , Obstrução da Artéria Renal/metabolismo , Animais , Pressão Sanguínea , Antígenos CD40/genética , Linhagem Celular , Modelos Animais de Doenças , Fibrose , Taxa de Filtração Glomerular , Humanos , Mediadores da Inflamação/metabolismo , Isquemia/genética , Isquemia/patologia , Isquemia/fisiopatologia , Rim/patologia , Rim/fisiopatologia , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Masculino , Estresse Oxidativo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ratos Endogâmicos Dahl , Obstrução da Artéria Renal/genética , Obstrução da Artéria Renal/patologia , Obstrução da Artéria Renal/fisiopatologia , Transdução de Sinais
7.
Am J Hypertens ; 33(8): 765-774, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32179886

RESUMO

BACKGROUND: Mitochondria modulate endothelial cell (EC) function, but may be damaged during renal disease. We hypothesized that the ischemic and metabolic constituents of swine renovascular disease (RVD) induce mitochondrial damage and impair the function of renal artery ECs. METHODS: Pigs were studied after 16 weeks of metabolic syndrome (MetS), renal artery stenosis (RAS), or MetS + RAS, and Lean pigs served as control (n = 6 each). Mitochondrial morphology, homeostasis, and function were measured in isolated primary stenotic-kidney artery ECs. EC functions were assessed in vitro, whereas vasoreactivity of renal artery segments was characterized in organ baths. RESULTS: Lean + RAS and MetS + RAS ECs showed increased mitochondrial area and decreased matrix density. Mitochondrial biogenesis was impaired in MetS and MetS + RAS compared with their respective controls. Mitochondrial membrane potential similarly decreased in MetS, Lean + RAS, and MetS + RAS groups, whereas production of reactive oxygen species increased in MetS vs. Lean, but further increased in both RAS groups. EC tube formation was impaired in MetS, RAS, and MetS + RAS vs. Lean, but EC proliferation and endothelial-dependent relaxation of renal artery segments were blunted in MetS vs. Lean, but further attenuated in Lean + RAS and MetS + RAS. CONCLUSIONS: MetS and RAS damage mitochondria in pig renal artery ECs, which may impair EC function. Coexisting MetS and RAS did not aggravate EC mitochondrial damage in the short time of our in vivo studies, suggesting that mitochondrial injury is associated with impaired renal artery EC function.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/fisiopatologia , Hipertensão Renovascular/metabolismo , Mitocôndrias/metabolismo , Artéria Renal/metabolismo , Vasodilatação/fisiologia , Animais , Modelos Animais de Doenças , Células Endoteliais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Feminino , Hipertensão Renovascular/patologia , Hipertensão Renovascular/fisiopatologia , Potencial da Membrana Mitocondrial/fisiologia , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Mitocôndrias/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Distribuição Aleatória , Espécies Reativas de Oxigênio/metabolismo , Artéria Renal/patologia , Artéria Renal/fisiopatologia , Obstrução da Artéria Renal/metabolismo , Obstrução da Artéria Renal/patologia , Sus scrofa , Suínos , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Cell Physiol Biochem ; 52(3): 617-632, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30907989

RESUMO

BACKGROUND/AIMS: Atherosclerotic renal artery stenosis (ARAS) may cause kidney injury and mitochondrial dysfunction, which is linked to cellular senescence. Elamipretide, a mitochondria-targeted peptide, improves renal function in ARAS, but whether it alleviates senescence is unknown. We hypothesized that elamipretide would reduce senescence stenotic kidney (STK) in ARAS. METHODS: Domestic pigs were randomized to control and unilateral ARAS untreated or treated with subcutaneous elamipretide (5d/wk) for 4 weeks starting after 6 weeks of ARAS or sham (n=6 each). After completion of treatment, STK renal blood flow (RBF) and glomerular filtration rate (GFR) were assessed in-vivo using multi-detector computed-tomography. Renal fibrosis and oxidative stress were analyzed in trichrome- and dihydroethidium-stained slides, respectively. Mitochondrial markers involved in the electrontransport chain (COX4, ATP/ADP ratio), biogenesis (PGC1α, PPARα), dynamics (MFN2, DRP1), and mitophagy (parkin, p62) were measured in the kidney using ELISA, western-blot, and immunohistochemistry. Cellular senescence (senescence-associated ß-galactosidase and heterochromatin foci, phosphorylated-H2AX, and p16/21/53) and senescence-associated secretory phenotype (SASP; PAI-1, MCP-1, TGFß, and TNFα) markers were studied by microscopy, quantitative reverse transcription-polymerase chain reaction, and western-blot. RESULTS: Blood pressure was elevated whereas STK-RBF and GFR were decreased in ARAS pigs, and tissue scarring was increased. ARAS induced STK cellular senescence and accumulated dysfunctional mitochondria, which were associated with cardiolipin loss, upregulated mitochondrial biogenesis, and defective mitophagy. Elamipretide normalized STK-RBF and GFR, alleviated fibrosis and oxidative stress, and restored mitochondrial cardiolipin, biogenesis, and mitophagy in ARAS, but did not change SASP markers, and attenuated only senescenceassociated ß-galactosidase activity and p53 gene expression. CONCLUSION: Mitochondrial protection improved renal function and fibrosis in the ARAS STK, but only partly mitigated cellular senescence. This finding suggests that mitochondrial dysfunction may not be a major determinant of cellular senescence in the early stage of ARAS.


Assuntos
Senescência Celular , Rim/fisiologia , Mitocôndrias/metabolismo , Obstrução da Artéria Renal/patologia , Animais , Cardiolipinas/metabolismo , Senescência Celular/efeitos dos fármacos , Creatinina/sangue , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Fibrose , Taxa de Filtração Glomerular , Rim/patologia , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Obstrução da Artéria Renal/tratamento farmacológico , Obstrução da Artéria Renal/metabolismo , Circulação Renal/efeitos dos fármacos , Suínos , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
9.
Am J Hypertens ; 31(12): 1307-1316, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30107490

RESUMO

BACKGROUND: The collateral circulation is important in maintenance of blood supply to the ischemic kidney distal to renal artery stenosis (RAS). Obesity metabolic syndrome (MetS) preserves renal blood flow (RBF) in the stenotic kidney, but whether this is related to an increase of collateral vessel growth is unknown. We hypothesized that MetS increased collateral circulation around the renal artery. METHODS: Twenty-one domestic pigs were randomly divided into unilateral RAS fed an atherogenic (high-fat/high-fructose, MetS-RAS) or standard diet, or controls (n = 7 each). RBF, glomerular filtration rate (GFR), and the peristenotic collateral circulation were assessed after 10 weeks using multidetector computed tomography (CT) and the intrarenal microcirculation by micro-CT. Vascular endothelial growth factor (VEGF) expression was studied in the renal artery wall, kidney, and perirenal fat. Renal fibrosis and stiffness were examined by trichrome and magnetic resonance elastography. RESULTS: Compared with controls, RBF and GFR were decreased in RAS, but not in MetS-RAS. MetS-RAS formed peristenotic collaterals to the same extent as RAS pigs but induced greater intrarenal microvascular loss, fibrosis, stiffness, and inflammation. MetS-RAS also attenuated VEGF expression in the renal tissue compared with RAS, despite increased expression in the perirenal fat. CONCLUSIONS: MetS does not interfere with collateral vessel formation in the stenotic kidney, possibly because decreased renal arterial VEGF expression offsets its upregulation in perirenal fat, arguing against a major contribution of the collateral circulation to preserve renal function in MetS-RAS. Furthermore, preserved renal function does not protect the poststenotic kidney from parenchymal injury.


Assuntos
Circulação Colateral , Rim/irrigação sanguínea , Síndrome Metabólica/fisiopatologia , Microcirculação , Obstrução da Artéria Renal/fisiopatologia , Artéria Renal/fisiopatologia , Circulação Renal , Animais , Velocidade do Fluxo Sanguíneo , Modelos Animais de Doenças , Rim/patologia , Síndrome Metabólica/complicações , Síndrome Metabólica/metabolismo , Artéria Renal/metabolismo , Obstrução da Artéria Renal/complicações , Obstrução da Artéria Renal/metabolismo , Obstrução da Artéria Renal/patologia , Sus scrofa , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Am J Physiol Renal Physiol ; 315(4): F747-F751, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29442546

RESUMO

Renovascular disease (RVD), which is prevalent in the elderly, significantly increases cardiovascular risk and can progressively deteriorate renal function. The loss of renal function in patients with RVD is associated with a progressive dysfunction, damage, and loss of renal microvessels, which can be combined with decreased renal bioavailability of vascular endothelial growth factor (VEGF) and a defective vascular repair and proliferation. This association has been the impetus for recent efforts that have focused on developing methods to stop the progression of renal injury by protecting the renal microvasculature. This mini-review focuses on recent studies supporting potential applications of VEGF therapy for the kidney and discusses underlying mechanisms of renoprotection.


Assuntos
Rim/metabolismo , Neovascularização Fisiológica/fisiologia , Circulação Renal/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Humanos , Rim/irrigação sanguínea , Obstrução da Artéria Renal/metabolismo
11.
Kidney Int ; 93(4): 842-854, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29273331

RESUMO

We recently developed a therapeutic biopolymer composed of an elastin-like polypeptide (ELP) fused to vascular endothelial growth factor (VEGF) and showed long-term renoprotective effects in experimental renovascular disease after a single intra-renal administration. Here, we sought to determine the specificity, safety, efficacy, and mechanisms of renoprotection of ELP-VEGF after systemic therapy in renovascular disease. We tested whether kidney selectivity of the ELP carrier would reduce off-target binding of VEGF in other organs. In vivo bio-distribution after systemic administration of ELP-VEGF in swine was determined in kidneys, liver, spleen, and heart. Stenotic-kidney renal blood flow and glomerular filtration rate were quantified in vivo using multi-detector computed tomography (CT) after six weeks of renovascular disease, then treated with a single intravenous dose of ELP-VEGF or placebo and observed for four weeks. CT studies were then repeated and the pigs euthanized. Ex vivo studies quantified renal microvascular density (micro-CT) and fibrosis. Kidneys, liver, spleen, and heart were excised to quantify the expression of angiogenic mediators and markers of progenitor cells. ELP-VEGF accumulated predominantly in the kidney and stimulated renal blood flow, glomerular filtration rate, improved cortical microvascular density, and renal fibrosis, and was accompanied by enhanced renal expression of VEGF, downstream mediators of VEGF signaling, and markers of progenitor cells compared to placebo. Expression of angiogenic factors in liver, spleen, and heart were not different compared to placebo-control. Thus, ELP efficiently directs VEGF to the kidney after systemic administration and induces long-term renoprotection without off-target effects, supporting the feasibility and safety of renal therapeutic angiogenesis via systemic administration of a novel kidney-specific bioengineered compound.


Assuntos
Indutores da Angiogênese/farmacologia , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Peptídeos/farmacologia , Obstrução da Artéria Renal/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/farmacologia , Indutores da Angiogênese/administração & dosagem , Indutores da Angiogênese/farmacocinética , Indutores da Angiogênese/toxicidade , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Modelos Animais de Doenças , Portadores de Fármacos , Fibrose , Taxa de Filtração Glomerular/efeitos dos fármacos , Injeções Intravenosas , Rim/metabolismo , Rim/patologia , Peptídeos/administração & dosagem , Peptídeos/farmacocinética , Peptídeos/toxicidade , Proteínas Recombinantes de Fusão/farmacologia , Obstrução da Artéria Renal/metabolismo , Obstrução da Artéria Renal/patologia , Obstrução da Artéria Renal/fisiopatologia , Circulação Renal/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Sus scrofa , Distribuição Tecidual , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/farmacocinética , Fator A de Crescimento do Endotélio Vascular/toxicidade
12.
Kidney Int ; 92(1): 114-124, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28242034

RESUMO

Mesenchymal stem/stromal cells (MSCs) have distinct capability for renal repair, but may have safety concerns. MSC-derived extracellular vesicles emerged as a novel noncellular alternative. Using a porcine model of metabolic syndrome and renal artery stenosis we tested whether extracellular vesicles attenuate renal inflammation, and if this capacity is mediated by their cargo of the anti-inflammatory cytokine interleukin (IL) 10. Pigs with metabolic syndrome were studied after 16 weeks of renal artery stenosis untreated or treated four weeks earlier with a single intrarenal delivery of extracellular vesicles harvested from adipose tissue-derived autologous MSCs. Lean and sham metabolic syndrome animals served as controls (seven each). Five additional pigs with metabolic syndrome and renal artery stenosis received extracellular vesicles with pre-silenced IL10 (IL10 knock-down). Single-kidney renal blood flow, glomerular filtration rate, and oxygenation were studied in vivo and renal injury pathways ex vivo. Retention of extracellular vesicles in the stenotic kidney peaked two days after delivery and decreased thereafter. Four weeks after injection, extracellular vesicle fragments colocalized with stenotic-kidney tubular cells and macrophages, indicating internalization or fusion. Extracellular vesicle delivery attenuated renal inflammation, and improved medullary oxygenation and fibrosis. Renal blood flow and glomerular filtration rate fell in metabolic syndrome and renal artery stenosis compared to metabolic syndrome, but was restored in pigs treated with extracellular vesicles. These renoprotective effects were blunted in pigs treated with IL10-depleted extracellular vesicles. Thus, extracellular vesicle-based regenerative strategies might be useful for patients with metabolic syndrome and renal artery stenosis.


Assuntos
Vesículas Extracelulares/transplante , Rim , Transplante de Células-Tronco Mesenquimais/métodos , Síndrome Metabólica/cirurgia , Nefrite/prevenção & controle , Obstrução da Artéria Renal/cirurgia , Animais , Hipóxia Celular , Células Cultivadas , Modelos Animais de Doenças , Vesículas Extracelulares/metabolismo , Feminino , Fibrose , Taxa de Filtração Glomerular , Interleucina-10/genética , Interleucina-10/metabolismo , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Síndrome Metabólica/complicações , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Nefrite/etiologia , Nefrite/genética , Nefrite/metabolismo , Oxigênio/sangue , Interferência de RNA , Obstrução da Artéria Renal/complicações , Obstrução da Artéria Renal/genética , Obstrução da Artéria Renal/metabolismo , Circulação Renal , Sus scrofa , Fatores de Tempo , Transplante Autólogo
13.
Curr Hypertens Rep ; 18(6): 45, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27130448

RESUMO

Renal artery stenosis (RAS) is increasingly encountered in clinical practice. The two most common etiologies are fibromuscular dysplasia (FMD) and atherosclerotic renal artery disease (ARAS), with the latter accounting for the vast majority of cases. Significant RAS activates the renin-angiotensin-aldosterone system and is associated with three major clinical syndromes: ischemic nephropathy, hypertension, and destabilizing cardiac syndromes. Over the past two decades, advancements in diagnostic and interventional techniques have led to improved detection and the widespread use of endovascular renal artery revascularization strategies in the management of ARAS. However, renal artery stenting for ARAS remains controversial. Although several studies have demonstrated some benefit with renal artery revascularization, this has not been to the extent anticipated or predicted. Moreover, these trials have significant flaws in their study design and are hampered with inherent bias which make their interpretation challenging. In this review, we evaluate the existing body of evidence and offer an approach to the management of patients with ARAS in light of the current literature. From the data provided, identification of subgroup of patients, namely, those with a hemodynamically significant RAS in the context of progressive renal insufficiency and/or deteriorating arterial hypertension, seems possible and may derive clinical benefit from ARAS stent revascularization. Appropriate patient selection is therefore the key and more robust studies are required.


Assuntos
Implante de Prótese Vascular , Hipertensão Renovascular , Obstrução da Artéria Renal/cirurgia , Artéria Renal , Stents , Implante de Prótese Vascular/instrumentação , Implante de Prótese Vascular/métodos , Humanos , Hipertensão Renovascular/etiologia , Hipertensão Renovascular/metabolismo , Hipertensão Renovascular/prevenção & controle , Rim/irrigação sanguínea , Rim/fisiopatologia , Seleção de Pacientes , Obstrução da Artéria Renal/complicações , Obstrução da Artéria Renal/metabolismo , Sistema Renina-Angiotensina/fisiologia
14.
Am J Physiol Renal Physiol ; 310(1): F6-9, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26538439

RESUMO

Renal artery stenosis is the main cause of renovascular hypertension and results in ischemic nephropathy characterized by inflammation, oxidative stress, microvascular loss, and fibrosis with consequent functional failure. Considering the limited number of strategies that effectively control renovascular hypertension and restore renal function, we propose that cell therapy may be a promising option based on the regenerative and immunosuppressive properties of stem cells. This review addresses the effects of mesenchymal stem cells (MSC) in an experimental animal model of renovascular hypertension known as 2 kidney-1 clip (2K-1C). Significant benefits of MSC treatment have been observed on blood pressure and renal structure of the stenotic kidney. The mechanisms involved are discussed.


Assuntos
Hipertensão Renovascular/cirurgia , Rim , Transplante de Células-Tronco Mesenquimais , Obstrução da Artéria Renal/cirurgia , Animais , Doença Crônica , Modelos Animais de Doenças , Humanos , Hipertensão Renovascular/imunologia , Hipertensão Renovascular/metabolismo , Hipertensão Renovascular/patologia , Hipertensão Renovascular/fisiopatologia , Rim/imunologia , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina , Recuperação de Função Fisiológica , Regeneração , Obstrução da Artéria Renal/imunologia , Obstrução da Artéria Renal/metabolismo , Obstrução da Artéria Renal/patologia , Obstrução da Artéria Renal/fisiopatologia , Transdução de Sinais
15.
Am J Physiol Renal Physiol ; 310(5): F372-84, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26661648

RESUMO

Renovascular hypertension (RVH) is a common cause of both cardiovascular and renal morbidity and mortality. In renal artery stenosis (RAS), atrophy in the stenotic kidney is associated with an influx of macrophages and other mononuclear cells. We tested the hypothesis that chemokine receptor 2 (CCR2) inhibition would reduce chronic renal injury by reducing macrophage influx in the stenotic kidney of mice with RAS. We employed a well-established murine model of RVH to define the relationship between macrophage infiltration and development of renal atrophy in the stenotic kidney. To determine the role of chemokine ligand 2 (CCL2)/CCR2 signaling in the development of renal atrophy, mice were treated with the CCR2 inhibitor RS-102895 at the time of RAS surgery and followed for 4 wk. Renal tubular epithelial cells expressed CCL2 by 3 days following surgery, a time at which no significant light microscopic alterations, including interstitial inflammation, were identified. Macrophage influx increased with time following surgery. At 4 wk, the development of severe renal atrophy was accompanied by an influx of inducible nitric oxide synthase (iNOS)+ and CD206+ macrophages that coexpressed F4/80, with a modest increase in macrophages coexpressing arginase 1 and F4/80. The CCR2 inhibitor RS-102895 attenuated renal atrophy and significantly reduced the number of dual-stained F4/80+ iNOS+ and F4/80+ CD206+ but not F4/80+ arginase 1+ macrophages. CCR2 inhibition reduces iNOS+ and CD206+ macrophage accumulation that coexpress F4/80 and renal atrophy in experimental renal artery stenosis. CCR2 blockade may provide a novel therapeutic approach to humans with RVH.


Assuntos
Benzoxazinas/farmacologia , Quimiocina CCL2/metabolismo , Hipertensão Renovascular/tratamento farmacológico , Rim/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Piperidinas/farmacologia , Substâncias Protetoras/farmacologia , Receptores CCR2/antagonistas & inibidores , Obstrução da Artéria Renal/tratamento farmacológico , Animais , Antígenos de Diferenciação/metabolismo , Arginase/metabolismo , Atrofia , Quimiocina CCL2/genética , Citoproteção , Modelos Animais de Doenças , Hipertensão Renovascular/genética , Hipertensão Renovascular/metabolismo , Hipertensão Renovascular/patologia , Rim/metabolismo , Rim/patologia , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Terapia de Alvo Molecular , Nefrite Intersticial/metabolismo , Nefrite Intersticial/patologia , Nefrite Intersticial/prevenção & controle , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores CCR2/metabolismo , Receptores de Superfície Celular/metabolismo , Obstrução da Artéria Renal/genética , Obstrução da Artéria Renal/metabolismo , Obstrução da Artéria Renal/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
16.
PLoS One ; 10(4): e0124123, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25884946

RESUMO

BACKGROUND: A close association exists between renal impairment (RI) and atrial fibrillation (AF) occurrence. Increased activity of the sympathetic nervous system (SNS) may contribute to the development of AF associated with RI. Renal denervation (RDN) decreases central sympathetic activity. OBJECTIVE: The main objective of the study was to explore the effects of RDN on AF occurrence and its possible mechanisms in beagles with RI. METHODS: Unilateral RI was induced in beagles by embolization of small branches of the renal artery in the right kidney using gelatin sponge granules in Model (n = 6) and RDN group (n = 6). The Sham group (n = 6) underwent the same procedure, except for embolization. Then animals in RDN group underwent radiofrequency ablation of the renal sympathetic nerve. Cardiac electrophysiological parameters, blood pressure, left ventricular end-diastolic pressure, and AF inducibility were investigated. The activity of the SNS, renin-angiotensin-aldosterone system (RAAS), inflammation and atrial interstitial fibrosis were measured. RESULTS: Embolization of small branches of the renal artery in the right kidney led to ischemic RI. Heart rate, P wave duration and BP were increased by RI, which were prevented or attenuated by RDN. Atrial effective refractory period was shortened and AF inducibility was increased by RI, which were prevented by RDN. Antegrade Wenckebach point was shortened, atrial and ventricular rates during AF were increased by RI, which were attenuated or prevented by RDN. Levels of norepinephrine, renin and aldosterone in plasma, norepinephrine, angiotensin II, aldosterone, interleukin-6 and high sensitivity C-reactive protein in atrial tissue were elevated, and atrial interstitial fibrosis was enhanced by RI, which were attenuated by RDN. CONCLUSIONS: RDN significantly reduced AF inducibility, prevented the atrial electrophysiological changes in a model of RI by combined reduction of sympathetic drive and RAAS activity, and inhibition of inflammation activity and fibrotic pathway in atrial tissue.


Assuntos
Fibrilação Atrial/prevenção & controle , Denervação Autônoma , Isquemia/cirurgia , Rim/inervação , Aldosterona/análise , Angiotensina II/análise , Animais , Fibrilação Atrial/etiologia , Fibrilação Atrial/metabolismo , Fibrilação Atrial/fisiopatologia , Denervação Autônoma/métodos , Proteína C-Reativa/análise , Ablação por Cateter , Modelos Animais de Doenças , Cães , Fibrose , Átrios do Coração/química , Átrios do Coração/patologia , Hemodinâmica , Interleucina-6/análise , Isquemia/complicações , Isquemia/metabolismo , Isquemia/fisiopatologia , Rim/irrigação sanguínea , Modelos Cardiovasculares , Norepinefrina/análise , Artéria Renal/patologia , Obstrução da Artéria Renal/complicações , Obstrução da Artéria Renal/metabolismo , Obstrução da Artéria Renal/fisiopatologia , Renina/sangue , Sistema Renina-Angiotensina/fisiologia , Sistema Nervoso Simpático/fisiopatologia
17.
Nephrol Dial Transplant ; 29(2): 274-82, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24097799

RESUMO

BACKGROUND: Renal parenchymal inflammation is a critical determinant of kidney injury in renal artery stenosis (RAS) but is difficult to assess in the single kidney without tissue samples. Whether renal vein (RV) levels of inflammatory markers reflect active parenchymal inflammation remains unknown. We evaluated the relationship between net RV cytokine release and tissue inflammation in the post-stenotic kidney. METHODS: Pigs were studied after 10 weeks of RAS treated 4 weeks earlier with intra-renal vehicle or anti-inflammatory mesenchymal stem cells (MSCs) or normal control. Single-kidney renal blood flow was measured by fast computerized tomography. RV and inferior vena cava levels of tumor necrosis factor (TNF)-α, interferon (IF)-γ, monocyte chemoattractant protein (MCP-1) and interleukin (IL)-10 were measured by enzyme-linked immunosorbent assay, and their net release calculated. Renal expression of the same cytokines was correlated with their net release. RESULTS: Net release of TNF-α, IF-γ and MCP-1 was higher in RAS compared with normal and to the contralateral kidney (all P<0.05), decreased in MSC-treated pigs as was their tissue expression. Contrarily, the release of the anti-inflammatory IL-10 was lower in RAS and normalized in RAS+MSC. The net release of TNF-α, MCP-1 and IL-10 directly correlated with their tissue expression. The ratio of inflammatory-to-reparative macrophages directly correlated with the release of MCP-1, but inversely with the release of IL-10. In vitro cultured MSCs also induced a shift in the macrophage phenotype from inflammatory (M1) to reparative (M2). CONCLUSIONS: Our findings demonstrate that the release of inflammatory markers from the affected kidney provides an index of renal tissue inflammation in experimental RAS.


Assuntos
Citocinas/metabolismo , Endotélio Vascular/metabolismo , Nefrite Intersticial/metabolismo , Obstrução da Artéria Renal/metabolismo , Veias Renais/fisiologia , Animais , Biomarcadores/metabolismo , Proliferação de Células , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Taxa de Filtração Glomerular , Interferon gama/metabolismo , Interleucina-10/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Células-Tronco Mesenquimais/patologia , Nefrite Intersticial/patologia , Estresse Oxidativo , Obstrução da Artéria Renal/complicações , Obstrução da Artéria Renal/fisiopatologia , Suínos , Fator de Necrose Tumoral alfa/metabolismo
18.
Stem Cells ; 31(1): 117-25, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23097349

RESUMO

Endothelial progenitor cells (EPC) and mesenchymal stem cells (MSC) augment tissue repair but possess slightly different properties. How the cellular phenotype affects the efficacy of this approach in renovascular disease is incompletely understood. This study tested the hypothesis that EPC and MSC protect the poststenotic kidney by blunting different disease pathways. Peripheral blood EPC and adipose-derived MSC were expanded and characterized by cell surface markers (e.g., CD34/kinase insert domain receptor, or CD44/CD90). Single-kidney hemodynamics and function were assessed in pigs after 10 weeks of renal artery stenosis (RAS) treated 4 weeks earlier with an intrarenal infusion of vehicle (n = 7), EPC (RAS+EPC) or MSC (RAS+MSC) (both 10 × 10(6), n = 6), and normal controls (n = 7). Kidney disease mechanisms were evaluated ex vivo. The ability of EPC and MSC to attenuate endoplasmic reticulum (ER) stress was also studied in isolated ER and in tubular cells cocultured with EPC and MSC. Glomerular filtration rate in RAS was lower than controls, increased in RAS+EPC, and further improved in RAS+MSC, although both improved renal blood flow similarly. EPC prominently enhanced renal growth factor expression and decreased oxidative stress, while MSC more significantly attenuated renal inflammation, ER stress, and apoptosis. Furthermore, MSC induced a greater decrease in caspase-3 and CHOP expression in cultured tubular cells through mechanisms involving cell contact. EPC and MSC achieve a comparable decrease of kidney injury in RAS by different mechanisms, although MSC elicited slightly superior improvement of renal function. These results support development of cell-based approaches for management of renovascular disease and suggest cell selection based on the underlying pathophysiology of kidney injury.


Assuntos
Injúria Renal Aguda/terapia , Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Obstrução da Artéria Renal/metabolismo , Animais , Antígenos CD34 , Apoptose , Caspase 3/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Estresse do Retículo Endoplasmático , Taxa de Filtração Glomerular , Receptores de Hialuronatos , Rim/irrigação sanguínea , Rim/metabolismo , Rim/patologia , Estresse Oxidativo , Obstrução da Artéria Renal/patologia , Circulação Renal , Suínos , Antígenos Thy-1
19.
Hypertension ; 60(5): 1242-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23045468

RESUMO

Revascularization improves blood pressure but not renal function in most patients with atherosclerotic renal artery stenosis (ARAS), possibly related to injury incurred during renal reperfusion. Bendavia, a novel tetrapeptide that inhibits mitochondrial permeability transition pore opening, reduces apoptosis, oxidative stress, and ischemia-reperfusion injury in experimental models. However, its potential for improving renal response to revascularization of chronic ARAS is unknown. We hypothesized that adjunct Bendavia would improve renal structure and function after percutaneous transluminal renal angioplasty (PTRA). Pigs were treated after 6 weeks of ARAS or control with PTRA+stenting (or sham), adjunct continuous 4-hour infusion of Bendavia (0.05 mg/kg IV) or vehicle (n=7 each) during PTRA. Single-kidney renal blood flow and glomerular filtration rate were studied 4 weeks later and renal mitochondrial biogenesis, microvascular architecture, and injurious pathways evaluated ex vivo. Monocyte chemoattractant protein-1 levels rose after PTRA, suggesting inflammatory injury. Bendavia did not immediately affect inflammatory cytokine levels, yet 4 weeks later, stenotic kidney renal blood flow and glomerular filtration rate both improved (44.00 ± 0.21% and 36.40 ± 10.21%, respectively) in ARAS+PTRA+Bendavia compared with ARAS+PTRA+vehicle. Renal mitochondrial biogenesis was restored after PTRA+Bendavia, and microvascular rarefaction, apoptosis, oxidative stress, tubular injury, and fibrosis decreased. Infusion of Bendavia during PTRA preserved mitochondrial biogenesis, renal hemodynamics, and function, and attenuated tissue injury in swine ARAS. Thus, functional mitochondrial injury during renal reperfusion may sustain renal inflammatory injury and limit kidney recovery after PTRA. Potent antiapoptotic and antioxidant effects provide Bendavia a novel therapeutic potential for improving kidney outcomes after PTRA in experimental ARAS.


Assuntos
Aterosclerose/terapia , Rim/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/antagonistas & inibidores , Oligopeptídeos/farmacologia , Obstrução da Artéria Renal/terapia , Angioplastia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Apoptose/efeitos dos fármacos , Aterosclerose/metabolismo , Western Blotting , Quimiocina CCL2/metabolismo , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Rim/irrigação sanguínea , Rim/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Estresse Oxidativo/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Obstrução da Artéria Renal/metabolismo , Circulação Renal/efeitos dos fármacos , Suínos , Fatores de Tempo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Am J Physiol Renal Physiol ; 302(11): F1478-85, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22419692

RESUMO

Renal artery stenosis (RAS) promotes microvascular rarefaction and fibrogenesis, which may eventuate in irreversible kidney injury. We have shown that percutaneous transluminal renal angioplasty (PTRA) or endothelial progenitor cells (EPC) improve renal cortical hemodynamics and function in the poststenotic kidney. The renal medulla is particularly sensitive to hypoxia, yet little is known about reversibility of medullary injury on restoration of renal blood flow. This study was designed to test the hypothesis that PTRA, with or without adjunct EPC delivery to the stenotic kidney, may improve medullary remodeling and tubular function. RAS was induced in 21 pigs using implantation of irritant coils, while another group served as normal controls (n = 7 each). Two RAS groups were then treated 6 wk later with PTRA or both PTRA and EPC. Four weeks later, medullary hemodynamics, microvascular architecture, and oxygen-dependent tubular function of the stenotic kidneys were examined using multidetector computed tomography, microcomputed tomography, and blood oxygenation level-dependent MRI, respectively. Medullary protein expression of vascular endothelial growth factor, endothelial nitric oxide synthase, hypoxia-inducible factor-1α, and NAD(P)H oxidase p47 were determined. All RAS groups showed decreased medullary vascular density and blood flow. However, in RAS+PTRA+EPC animals, EPC were engrafted in tubular structures, oxygen-dependent tubular function was normalized, and fibrosis attenuated, despite elevated expression of hypoxia-inducible factor-1α and sustained downregulation of vascular endothelial growth factor. In conclusion, EPC delivery, in addition to PTRA, restores medullary oxygen-dependent tubular function, despite impaired medullary blood and oxygen supply. These results support further development of cell-based therapy as an adjunct to revascularization of RAS.


Assuntos
Células Endoteliais/transplante , Transplante de Células-Tronco Hematopoéticas , Medula Renal/metabolismo , Túbulos Renais/metabolismo , Consumo de Oxigênio/fisiologia , Obstrução da Artéria Renal/metabolismo , Circulação Renal/fisiologia , Animais , Volume Sanguíneo/fisiologia , Células Endoteliais/fisiologia , Feminino , Fibrose , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Processamento de Imagem Assistida por Computador , Rim/patologia , Medula Renal/patologia , Túbulos Renais/patologia , Imageamento por Ressonância Magnética , Microvasos/patologia , Oxigênio/sangue , Proteinúria/metabolismo , Obstrução da Artéria Renal/patologia , Suínos , Tomografia Computadorizada por Raios X , Fator A de Crescimento do Endotélio Vascular/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA