Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int J Obes (Lond) ; 46(6): 1222-1233, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35256761

RESUMEN

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.


Asunto(s)
Células Madre Mesenquimatosas , Obstrucción de la Arteria Renal , Animales , Humanos , Riñón/patología , Células Madre Mesenquimatosas/metabolismo , Ratones , Obesidad/metabolismo , Obstrucción de la Arteria Renal/metabolismo , Obstrucción de la Arteria Renal/patología , Factor A de Crecimiento Endotelial Vascular
2.
Am J Physiol Renal Physiol ; 320(3): F454-F463, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33554782

RESUMEN

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.


Asunto(s)
Células Epiteliales/citología , Macrófagos/citología , Células Madre Mesenquimatosas/citología , Fenotipo , Obstrucción de la Arteria Renal/patología , Animales , Técnicas de Cocultivo , Citocinas/metabolismo , Humanos , Riñón/metabolismo , Riñón/patología , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/metabolismo , Sustancias Protectoras/farmacología , Obstrucción de la Arteria Renal/metabolismo , Porcinos , Factor de Necrosis Tumoral alfa/metabolismo
3.
Am J Physiol Renal Physiol ; 319(1): F19-F28, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32463728

RESUMEN

Scattered tubular-like cells (STCs) are dedifferentiated surviving tubular epithelial cells that repair neighboring injured cells. Experimental renal artery stenosis (RAS) impairs STC reparative potency by inducing mitochondrial injury, but the exact mechanisms of mitochondrial damage remain unknown. We hypothesized that RAS alters expression of mitochondria-related genes, contributing to mitochondrial structural damage and dysfunction in swine STCs. CD24+/CD133+ STCs were isolated from pig kidneys after 10 wk of RAS or sham (n = 3 each). mRNA sequencing was performed, and nuclear DNA (nDNA)-encoded mitochondrial genes and mitochondrial DNA (mtDNA)-encoded genes were identified. Mitochondrial structure, ATP generation, biogenesis, and expression of mitochondria-associated microRNAs were also assessed. There were 96 nDNA-encoded mitochondrial genes upregulated and 12 mtDNA-encoded genes downregulated in RAS-STCs versus normal STCs. Functional analysis revealed that nDNA-encoded and mtDNA-encoded differentially expressed genes were primarily implicated in mitochondrial respiration and ATP synthesis. Mitochondria from RAS STCs were swollen and showed cristae remodeling and loss and decreased ATP production. Immunoreactivity of the mitochondrial biogenesis marker peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α and expression of the mitochondria-associated microRNAs miR-15a, miR-181a, miR-196a, and miR-296-3p, which target several mtDNA genes, were higher in RAS-STCs compared with normal STCs, suggesting a potential modulation of mitochondria-related gene expression. These results demonstrate that RAS induces an imbalance in mtDNA- and nDNA-mitochondrial gene expression, impairing mitochondrial structure and function in swine STCs. These observations support development of gene gain- and loss-of-function strategies to ameliorate mitochondrial damage and preserve the reparative potency of STCs in patients with renal ischemia.


Asunto(s)
Expresión Génica , Genes Mitocondriales , Isquemia/genética , Riñón/irrigación sanguínea , Mitocondrias/metabolismo , Obstrucción de la Arteria Renal/metabolismo , Animales , Femenino , Isquemia/metabolismo , Biogénesis de Organelos , Obstrucción de la Arteria Renal/genética , Porcinos
4.
Cell Physiol Biochem ; 52(3): 617-632, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30907989

RESUMEN

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.


Asunto(s)
Senescencia Celular , Riñón/fisiología , Mitocondrias/metabolismo , Obstrucción de la Arteria Renal/patología , Animales , Cardiolipinas/metabolismo , Senescencia Celular/efectos de los fármacos , Creatinina/sangre , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Fibrosis , Tasa de Filtración Glomerular , Riñón/patología , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Obstrucción de la Arteria Renal/tratamiento farmacológico , Obstrucción de la Arteria Renal/metabolismo , Circulación Renal/efectos de los fármacos , Porcinos , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
5.
Int J Mol Sci ; 20(20)2019 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-31614781

RESUMEN

BACKGROUND: Scattered tubular-like cells (STCs) proliferate and differentiate to support neighboring injured renal tubular cells during recovery from insults. Renal artery stenosis (RAS) induces renal ischemia and hypertension and leads to loss of kidney function, but whether RAS alters renal endogenous repair mechanisms, such as STCs, remains unknown. We hypothesize that RAS in swine modifies the messenger RNA (mRNA) profile of STCs, blunting their in vitro reparative capacity. METHODS: CD24+/CD133+ STCs were isolated from pig kidneys after 10-weeks of RAS or sham (n = 3 each) and their gene cargo analyzed using high-throughput mRNAseq. Expression profiles for upregulated and downregulated mRNAs in RAS-STCs were functionally interpreted by gene ontology analysis. STC activation was assessed by counting the total number of STCs in pig kidney sections using flow cytometry, whereas cell proliferation was assessed in vitro. RESULTS: Of all expressed genes, 1430 genes were upregulated and 315 downregulated in RAS- versus Normal-STCs. Expression of selected candidate genes followed the same fold change directions as the mRNAseq findings. Genes upregulated in RAS-STCs were involved in cell adhesion, extracellular matrix remodeling, and kidney development, whereas those downregulated in RAS-STCs are related to cell cycle and cytoskeleton. The percentage of STCs from dissociated kidney cells was higher in RAS versus Normal pigs, but their proliferation rate was blunted. CONCLUSIONS: Renal ischemia and hypertension in swine induce changes in the mRNA profile of STCs, associated with increased STC activation and impaired proliferation. These observations suggest that RAS may alter the reparative capacity of STCs.


Asunto(s)
Obstrucción de la Arteria Renal/genética , Transcriptoma , Animales , Células Cultivadas , Femenino , Túbulos Renales/citología , Túbulos Renales/metabolismo , Obstrucción de la Arteria Renal/metabolismo , Porcinos
6.
Am J Physiol Renal Physiol ; 315(4): F747-F751, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29442546

RESUMEN

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.


Asunto(s)
Riñón/metabolismo , Neovascularización Fisiológica/fisiología , Circulación Renal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Humanos , Riñón/irrigación sanguínea , Obstrucción de la Arteria Renal/metabolismo
7.
Kidney Int ; 93(4): 842-854, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29273331

RESUMEN

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.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Péptidos/farmacología , Obstrucción de la Arteria Renal/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/farmacología , Inductores de la Angiogénesis/administración & dosificación , Inductores de la Angiogénesis/farmacocinética , Inductores de la Angiogénesis/toxicidad , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Portadores de Fármacos , Fibrosis , Tasa de Filtración Glomerular/efectos de los fármacos , Inyecciones Intravenosas , Riñón/metabolismo , Riñón/patología , Péptidos/administración & dosificación , Péptidos/farmacocinética , Péptidos/toxicidad , Proteínas Recombinantes de Fusión/farmacología , Obstrucción de la Arteria Renal/metabolismo , Obstrucción de la Arteria Renal/patología , Obstrucción de la Arteria Renal/fisiopatología , Circulación Renal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Sus scrofa , Distribución Tisular , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/farmacocinética , Factor A de Crecimiento Endotelial Vascular/toxicidad
8.
Kidney Int ; 92(1): 114-124, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28242034

RESUMEN

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.


Asunto(s)
Vesículas Extracelulares/trasplante , Riñón , Trasplante de Células Madre Mesenquimatosas/métodos , Síndrome Metabólico/cirugía , Nefritis/prevención & control , Obstrucción de la Arteria Renal/cirugía , Animales , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Femenino , Fibrosis , Tasa de Filtración Glomerular , Interleucina-10/genética , Interleucina-10/metabolismo , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Síndrome Metabólico/complicaciones , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , Nefritis/etiología , Nefritis/genética , Nefritis/metabolismo , Oxígeno/sangre , Interferencia de ARN , Obstrucción de la Arteria Renal/complicaciones , Obstrucción de la Arteria Renal/genética , Obstrucción de la Arteria Renal/metabolismo , Circulación Renal , Sus scrofa , Factores de Tiempo , Trasplante Autólogo
9.
Am J Physiol Renal Physiol ; 310(1): F6-9, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26538439

RESUMEN

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.


Asunto(s)
Hipertensión Renovascular/cirugía , Riñón , Trasplante de Células Madre Mesenquimatosas , Obstrucción de la Arteria Renal/cirugía , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Hipertensión Renovascular/inmunología , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/patología , Hipertensión Renovascular/fisiopatología , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina , Recuperación de la Función , Regeneración , Obstrucción de la Arteria Renal/inmunología , Obstrucción de la Arteria Renal/metabolismo , Obstrucción de la Arteria Renal/patología , Obstrucción de la Arteria Renal/fisiopatología , Transducción de Señal
10.
Am J Physiol Renal Physiol ; 310(5): F372-84, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26661648

RESUMEN

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.


Asunto(s)
Benzoxazinas/farmacología , Quimiocina CCL2/metabolismo , Hipertensión Renovascular/tratamiento farmacológico , Riñón/efectos de los fármacos , Macrófagos/efectos de los fármacos , Piperidinas/farmacología , Sustancias Protectoras/farmacología , Receptores CCR2/antagonistas & inhibidores , Obstrucción de la Arteria Renal/tratamiento farmacológico , Animales , Antígenos de Diferenciación/metabolismo , Arginasa/metabolismo , Atrofia , Quimiocina CCL2/genética , Citoprotección , Modelos Animales de Enfermedad , Hipertensión Renovascular/genética , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/patología , Riñón/metabolismo , Riñón/patología , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Nefritis Intersticial/metabolismo , Nefritis Intersticial/patología , Nefritis Intersticial/prevención & control , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores CCR2/metabolismo , Receptores de Superficie Celular/metabolismo , Obstrucción de la Arteria Renal/genética , Obstrucción de la Arteria Renal/metabolismo , Obstrucción de la Arteria Renal/patología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
11.
Curr Hypertens Rep ; 18(6): 45, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27130448

RESUMEN

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.


Asunto(s)
Implantación de Prótesis Vascular , Hipertensión Renovascular , Obstrucción de la Arteria Renal/cirugía , Arteria Renal , Stents , Implantación de Prótesis Vascular/instrumentación , Implantación de Prótesis Vascular/métodos , Humanos , Hipertensión Renovascular/etiología , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/prevención & control , Riñón/irrigación sanguínea , Riñón/fisiopatología , Selección de Paciente , Obstrucción de la Arteria Renal/complicaciones , Obstrucción de la Arteria Renal/metabolismo , Sistema Renina-Angiotensina/fisiología
12.
Am J Physiol Renal Physiol ; 309(4): F273-9, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26041447

RESUMEN

Obesity remains a prominent public health concern. Obesity not only contributes greatly to cardiovascular events but has also been identified to initiate and affect the progression of preexisting chronic kidney disease. The prevalence of renal artery stenosis is growing world-wide, especially in the elderly population and in individuals with atherosclerotic risk factors such as obesity. Prolonged renovascular disease causes inflammation and microvascular remodeling within the post-stenotic kidney, which promote tissue scarring and may account for irreversible renal damage. Obesity has been shown to aggravate kidney damage via several pathways, including exacerbation of microvascular regression and renal cell injury mediated by adipocytes and insulin resistance, thereby worsening the structural and functional outcomes of the kidney in renovascular disease. Dietary modification and inhibition of the renin-angiotensin-aldosterone system have been shown to alleviate obesity-induced tissue injury and remodeling. Possibly, angiogenic factors may boost microvascular repair in the ischemic kidney in the obesity milieu. Novel therapeutic interventions targeting deleterious pathways that are activated by obesity and responsible for kidney damage need to be explored in future studies.


Asunto(s)
Hipertensión Renovascular , Obesidad , Obstrucción de la Arteria Renal , Insuficiencia Renal Crónica , Proteínas Angiogénicas/metabolismo , Animales , Hemodinámica , Humanos , Hipertensión Renovascular/epidemiología , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/fisiopatología , Hipertensión Renovascular/terapia , Microcirculación , Obesidad/epidemiología , Obesidad/metabolismo , Obesidad/fisiopatología , Obesidad/terapia , Prevalencia , Pronóstico , Obstrucción de la Arteria Renal/epidemiología , Obstrucción de la Arteria Renal/metabolismo , Obstrucción de la Arteria Renal/fisiopatología , Obstrucción de la Arteria Renal/terapia , Circulación Renal , Insuficiencia Renal Crónica/epidemiología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/terapia , Sistema Renina-Angiotensina , Medición de Riesgo , Factores de Riesgo , Transducción de Señal
13.
Kidney Int ; 85(4): 833-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24352153

RESUMEN

We hypothesized that chronic specific endothelin-A (ET-A) receptor blockade therapy would reverse renal dysfunction and injury in advanced experimental renovascular disease. To test this, unilateral renovascular disease was induced in 19 pigs, and after 6 weeks, single-kidney hemodynamics and function was quantified in vivo using computed tomography. All pigs with renovascular disease were divided such that seven were untreated, seven were treated with ET-A blockers, and five were treated with ET-B blockers. Four weeks later, all pigs were restudied in vivo, and then killed and ex vivo studies performed on the stenotic kidney to quantify microvascular density, remodeling, renal oxidative stress, inflammation, and fibrosis. Renal blood flow, glomerular filtration rate, and redox status were significantly improved in the stenotic kidney after ET-A but not ET-B blockade. Furthermore, only ET-A blockade therapy reversed renal microvascular rarefaction and diminished remodeling, which was accompanied by a marked decreased in renal inflammatory and fibrogenic activity. Thus, ET-A but not ET-B blockade ameliorated renal injury in pigs with advanced renovascular disease by stimulating microvascular proliferation and decreasing the progression of microvascular remodeling, renal inflammation, and fibrosis in the stenotic kidney. These effects were functionally consequential as ET-A blockade improved single kidney microvascular endothelial function, renal blood flow, and glomerular filtration rate, and decreased albuminuria.


Asunto(s)
Antagonistas de los Receptores de la Endotelina A/uso terapéutico , Antagonistas de los Receptores de la Endotelina B/uso terapéutico , Hipertensión Renovascular/tratamiento farmacológico , Obstrucción de la Arteria Renal/tratamiento farmacológico , Circulación Renal/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Antagonistas de los Receptores de la Endotelina A/farmacología , Antagonistas de los Receptores de la Endotelina B/farmacología , Endotelina-1/sangre , Hipertensión Renovascular/metabolismo , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Riñón/metabolismo , Microvasos/efectos de los fármacos , Tomografía Computarizada Multidetector , Obstrucción de la Arteria Renal/metabolismo , Porcinos
14.
Curr Opin Nephrol Hypertens ; 23(6): 525-32, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25160077

RESUMEN

PURPOSE OF REVIEW: The neutral findings of Angioplasty and Stenting for Renal Artery Lesions and Cardiovascular Outcomes in Renal Artery Lesions trials have shown that unselected revascularization does not improve outcomes in atherosclerotic renovascular disease (ARVD). This review highlights recent translational, clinical and epidemiological studies and suggests directions for future research. RECENT FINDINGS: Imaging studies show that the degree of renal artery stenosis is not the most important determinant of outcome and response to therapies in ARVD. Porcine models have established a better understanding of the microvascular and inflammatory changes that occur in ARVD. Biomarkers of inflammation and cardiovascular dysfunction may be informative but do not yet help assess prognosis or response to treatment. Stem cell therapies show promise in animal models but have yet to translate into clinical practice. Analysis of patient subgroups with high-risk presentations of ARVD has provided new insights into treatment response and may guide future studies. SUMMARY: It is time to reframe thinking and research in ARVD. We need better ways to identify patients likely to benefit from revascularization and to improve response to treatment in these individuals. Many preclinical studies show promise, but these are often small scale and difficult to replicate. Future work should focus on establishing an international disease registry as a foundation for collaborative research.


Asunto(s)
Angioplastia/instrumentación , Ensayos Clínicos como Asunto , Obstrucción de la Arteria Renal/terapia , Stents , Angioplastia/efectos adversos , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Medicina Basada en la Evidencia , Humanos , Selección de Paciente , Obstrucción de la Arteria Renal/diagnóstico , Obstrucción de la Arteria Renal/epidemiología , Obstrucción de la Arteria Renal/metabolismo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
15.
Stem Cells ; 31(1): 117-25, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097349

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/terapia , Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Obstrucción de la Arteria Renal/metabolismo , Animales , Antígenos CD34 , Apoptosis , Caspasa 3/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Estrés del Retículo Endoplásmico , Tasa de Filtración Glomerular , Receptores de Hialuranos , Riñón/irrigación sanguínea , Riñón/metabolismo , Riñón/patología , Estrés Oxidativo , Obstrucción de la Arteria Renal/patología , Circulación Renal , Porcinos , Antígenos Thy-1
16.
Nephrol Dial Transplant ; 29(2): 274-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24097799

RESUMEN

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.


Asunto(s)
Citocinas/metabolismo , Endotelio Vascular/metabolismo , Nefritis Intersticial/metabolismo , Obstrucción de la Arteria Renal/metabolismo , Venas Renales/fisiología , Animales , Biomarcadores/metabolismo , Proliferación Celular , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Tasa de Filtración Glomerular , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Células Madre Mesenquimatosas/patología , Nefritis Intersticial/patología , Estrés Oxidativo , Obstrucción de la Arteria Renal/complicaciones , Obstrucción de la Arteria Renal/fisiopatología , Porcinos , Factor de Necrosis Tumoral alfa/metabolismo
17.
Eur Heart J ; 34(7): 540-548a, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22771675

RESUMEN

AIMS: The mechanisms mediating kidney injury and repair in humans with atherosclerotic renal artery stenosis (ARAS) remain poorly understood. We hypothesized that the stenotic kidney releases inflammatory mediators and recruits progenitor cells to promote regeneration. METHODS AND RESULTS: Essential hypertensive (EH) and ARAS patients (n=24 each) were studied during controlled sodium intake and antihypertensive treatment. Inferior vena cava (IVC) and renal vein (RV) levels of CD34+/KDR+ progenitor cells, cell adhesion molecules, inflammatory biomarkers, progenitor cell homing signals, and pro-angiogenic factors were measured in EH and ARAS, and their gradient and net release compared with systemic levels in matched normotensive controls (n= 24). Blood pressure in ARAS was similar to EH, but the glomerular filtration rate was lower. Renal vein levels of soluble E-Selectin, vascular cell adhesion molecule-1, and several inflammatory markers were higher in the stenotic kidney RV vs. normal and EH RV (P < 0.05), and their net release increased. Similarly, stem-cell homing factor levels increased in the stenotic kidney RV. Systemic CD34+/KDR+ progenitor cell levels were lower in both EH and ARAS and correlated with cytokine levels. Moreover, CD34+/KDR+ progenitor cells developed a negative gradient across the ARAS kidney, suggesting progenitor cell retention. The non-stenotic kidney also showed signs of inflammatory processes, which were more subtle than in the stenotic kidney. CONCLUSION: Renal vein blood from post-stenotic human kidneys has multiple markers reflecting active inflammation that portends kidney injury and reduced function. CD34+/KDR+ progenitor cells sequestered within these kidneys may participate in reparative processes. These inflammation-related pathways and limited circulating progenitor cells may serve as novel therapeutic targets to repair the stenotic kidney.


Asunto(s)
Lesión Renal Aguda/metabolismo , Aterosclerosis/metabolismo , Hipertensión Renovascular/metabolismo , Obstrucción de la Arteria Renal/metabolismo , Células Madre/fisiología , Lesión Renal Aguda/fisiopatología , Anciano , Antígenos CD34/metabolismo , Arteritis/metabolismo , Aterosclerosis/fisiopatología , Biomarcadores/metabolismo , Presión Sanguínea/fisiología , Estudios de Casos y Controles , Citocinas/metabolismo , Selectina E/metabolismo , Femenino , Tasa de Filtración Glomerular/fisiología , Hemodinámica/fisiología , Humanos , Hipertensión Renovascular/fisiopatología , Riñón/irrigación sanguínea , Riñón/metabolismo , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Obstrucción de la Arteria Renal/fisiopatología , Venas Renales/metabolismo , Factor de Células Madre/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Vena Cava Inferior/metabolismo
18.
Am J Physiol Renal Physiol ; 302(11): F1478-85, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22419692

RESUMEN

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.


Asunto(s)
Células Endoteliales/trasplante , Trasplante de Células Madre Hematopoyéticas , Médula Renal/metabolismo , Túbulos Renales/metabolismo , Consumo de Oxígeno/fisiología , Obstrucción de la Arteria Renal/metabolismo , Circulación Renal/fisiología , Animales , Volumen Sanguíneo/fisiología , Células Endoteliales/fisiología , Femenino , Fibrosis , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Procesamiento de Imagen Asistido por Computador , Riñón/patología , Médula Renal/patología , Túbulos Renales/patología , Imagen por Resonancia Magnética , Microvasos/patología , Oxígeno/sangre , Proteinuria/metabolismo , Obstrucción de la Arteria Renal/patología , Porcinos , Tomografía Computarizada por Rayos X , Factor A de Crecimiento Endotelial Vascular/biosíntesis
19.
Am J Physiol Renal Physiol ; 302(5): F519-25, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22114209

RESUMEN

In this study, we compared the traditional murine model with renal pedicle clamp with models that clamped the renal artery or vein alone as well as to a whole body ischemia-reperfusion injury (WBIRI) model. Male C57BL/6J mice underwent either clamping of the renal artery, vein, or both (whole pedicle) for 30 or 45 min followed by reperfusion, or 10 min of cardiac arrest followed by resuscitation up to 24 h. After 30 min of ischemia, the mice with renal vein clamping showed the mostly increased serum creatinine and the most severe renal tubule injury. After 45 min of ischemia, all mice with renal vasculature clamping had a comparable increase in serum creatinine but the renal tubule injury was most severe in renal artery-clamped mice. Renal arterial blood flow was most decreased in mice with a renal vein clamp compared with a renal artery or pedicle clamp. A 30-or 45-min renal ischemia time led to a significant increase in the protein level of interleukin-6, keratinocyte-derived chemokine (KC), and granular colony-stimulating factor in the ischemic kidney, but the KC was the highest in the renal pedicle-clamped kidney and the lowest in the renal vein-clamped kidney. Of note, 10 min of WBIRI led to kidney dysfunction and structural injury, although less than longer time clamping of isolated renal vasculature. Our data demonstrate important differences in ischemic AKI models. Understanding these differences is important in designing future experimental studies in mice as well as clinical trials in humans.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Isquemia/fisiopatología , Riñón/irrigación sanguínea , Obstrucción de la Arteria Renal/fisiopatología , Arteria Renal/fisiopatología , Venas Renales/fisiopatología , Lesión Renal Aguda/metabolismo , Animales , Velocidad del Flujo Sanguíneo , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Isquemia/metabolismo , Riñón/metabolismo , Riñón/fisiopatología , Pruebas de Función Renal , Masculino , Ratones , Arteria Renal/metabolismo , Obstrucción de la Arteria Renal/metabolismo , Venas Renales/metabolismo
20.
Pediatr Nephrol ; 27(6): 1037-40, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22366877

RESUMEN

BACKGROUND: Hyponatremic hypertensive syndrome (HHS) is characterized by unilateral renal artery stenosis with secondary hypertension and glomerular and tubular dysfunction due to hyperfiltration and activation of the renin-angiotensin system (RAS). CASE-DIAGNOSIS/TREATMENT: We describe four children with HHS. All presented with polyuria and polydipsia, electrolyte disturbances, metabolic alkalosis, variable tubular dysfunction, and nephrotic range proteinuria along with hypertension. Interestingly, in one patient, glomerular and tubular abnormalities preceded the development of hypertension. All symptoms resolved after the underlying renal ischemia was corrected by percutaneous angioplasty. CONCLUSION: Hyponatremic hypertensive syndrome may be more common in children than previously thought. Clinicians should be alert of the signs and symptoms because cure is possible with timely diagnosis and treatment.


Asunto(s)
Hipertensión Renovascular/etiología , Hiponatremia/etiología , Obstrucción de la Arteria Renal/complicaciones , Angioplastia , Biomarcadores/sangre , Biomarcadores/orina , Preescolar , Femenino , Hemodinámica , Humanos , Hipertensión Renovascular/diagnóstico , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/fisiopatología , Hipertensión Renovascular/terapia , Hiponatremia/diagnóstico , Hiponatremia/metabolismo , Hiponatremia/fisiopatología , Hiponatremia/terapia , Lactante , Masculino , Obstrucción de la Arteria Renal/diagnóstico , Obstrucción de la Arteria Renal/metabolismo , Obstrucción de la Arteria Renal/fisiopatología , Obstrucción de la Arteria Renal/terapia , Sistema Renina-Angiotensina , Síndrome , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA