Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
2.
J Clin Invest ; 134(1)2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-37906287

RESUMEN

Mineralocorticoid excess commonly leads to hypertension (HTN) and kidney disease. In our study, we used single-cell expression and chromatin accessibility tools to characterize the mineralocorticoid target genes and cell types. We demonstrated that mineralocorticoid effects were established through open chromatin and target gene expression, primarily in principal and connecting tubule cells and, to a lesser extent, in segments of the distal convoluted tubule cells. We examined the kidney-protective effects of steroidal and nonsteroidal mineralocorticoid antagonists (MRAs), as well as of amiloride, an epithelial sodium channel inhibitor, in a rat model of deoxycorticosterone acetate, unilateral nephrectomy, and high-salt consumption-induced HTN and cardiorenal damage. All antihypertensive therapies protected against cardiorenal damage. However, finerenone was particularly effective in reducing albuminuria and improving gene expression changes in podocytes and proximal tubule cells, even with an equivalent reduction in blood pressure. We noted a strong correlation between the accumulation of injured/profibrotic tubule cells expressing secreted posphoprotein 1 (Spp1), Il34, and platelet-derived growth factor subunit b (Pdgfb) and the degree of fibrosis in rat kidneys. This gene signature also showed a potential for classifying human kidney samples. Our multiomics approach provides fresh insights into the possible mechanisms underlying HTN-associated kidney disease, the target cell types, the protective effects of steroidal and nonsteroidal MRAs, and amiloride.


Asunto(s)
Hipertensión , Enfermedades Renales , Ratas , Humanos , Animales , Antagonistas de Receptores de Mineralocorticoides/farmacología , Cromatina/genética , Amilorida/farmacología , Mineralocorticoides/farmacología , Riñón , Enfermedades Renales/genética , Perfilación de la Expresión Génica
3.
Nat Commun ; 14(1): 6531, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37848446

RESUMEN

Adiponectin is a secretory protein, primarily produced in adipocytes. However, low but detectable expression of adiponectin can be observed in cell types beyond adipocytes, particularly in kidney tubular cells, but its local renal role is unknown. We assessed the impact of renal adiponectin by utilizing male inducible kidney tubular cell-specific adiponectin overexpression or knockout mice. Kidney-specific adiponectin overexpression induces a doubling of phosphoenolpyruvate carboxylase expression and enhanced pyruvate-mediated glucose production, tricarboxylic acid cycle intermediates and an upregulation of fatty acid oxidation (FAO). Inhibition of FAO reduces the adiponectin-induced enhancement of glucose production, highlighting the role of FAO in the induction of renal gluconeogenesis. In contrast, mice lacking adiponectin in the kidney exhibit enhanced glucose tolerance, lower utilization and greater accumulation of lipid species. Hence, renal adiponectin is an inducer of gluconeogenesis by driving enhanced local FAO and further underlines the important systemic contribution of renal gluconeogenesis.


Asunto(s)
Adiponectina , Gluconeogénesis , Riñón , Animales , Masculino , Ratones , Adiponectina/genética , Adiponectina/metabolismo , Gluconeogénesis/genética , Gluconeogénesis/fisiología , Glucosa/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Ratones Noqueados , Ácido Pirúvico/metabolismo
4.
J Am Soc Nephrol ; 34(11): 1843-1862, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37639336

RESUMEN

SIGNIFICANCE STATEMENT: Mouse models have been widely used to understand kidney disease pathomechanisms and play an important role in drug discovery. However, these models have not been systematically analyzed and compared. The authors characterized 18 different mouse kidney disease models at both bulk and single-cell gene expression levels and compared single-cell gene expression data from diabetic kidney disease (DKD) mice and from patients with DKD. Although single cell-level gene expression changes were mostly model-specific, different disease models showed similar changes when compared at a pathway level. The authors also found that changes in fractions of cell types are major drivers of bulk gene expression differences. Although the authors found only a small overlap of single cell-level gene expression changes between the mouse DKD model and patients, they observed consistent pathway-level changes. BACKGROUND: Mouse models have been widely used to understand kidney disease pathomechanisms and play an important role in drug discovery. However, these models have not been systematically analyzed and compared. METHODS: We analyzed single-cell RNA sequencing data (36 samples) and bulk gene expression data (42 samples) from 18 commonly used mouse kidney disease models. We compared single-nucleus RNA sequencing data from a mouse diabetic kidney disease model with data from patients with diabetic kidney disease and healthy controls. RESULTS: We generated a uniformly processed mouse single-cell atlas containing information for nearly 300,000 cells, identifying all major kidney cell types and states. Our analysis revealed that changes in fractions of cell types are major drivers of differences in bulk gene expression. Although gene expression changes at the single-cell level were mostly model-specific, different disease models showed similar changes when compared at a pathway level. Tensor decomposition analysis highlighted the important changes in proximal tubule cells in disease states. Specifically, we identified important alterations in expression of metabolic and inflammation-associated pathways. The mouse diabetic kidney disease model and patients with diabetic kidney disease shared only a small number of conserved cell type-specific differentially expressed genes, but we observed pathway-level activation patterns conserved between mouse and human diabetic kidney disease samples. CONCLUSIONS: This study provides a comprehensive mouse kidney single-cell atlas and defines gene expression commonalities and differences in disease states in mice. The results highlight the key role of cell heterogeneity in driving changes in bulk gene expression and the limited overlap of single-cell gene expression changes between animal models and patients, but they also reveal consistent pathway-level changes.


Asunto(s)
Nefropatías Diabéticas , Humanos , Ratones , Animales , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Riñón/metabolismo , Túbulos Renales Proximales/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo
5.
Cell Rep Med ; 4(4): 100992, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37023747

RESUMEN

Diabetic kidney disease (DKD) is the most common cause of renal failure. Therapeutics development is hampered by our incomplete understanding of animal models on a cellular level. We show that ZSF1 rats recapitulate human DKD on a phenotypic and transcriptomic level. Tensor decomposition prioritizes proximal tubule (PT) and stroma as phenotype-relevant cell types exhibiting a continuous lineage relationship. As DKD features endothelial dysfunction, oxidative stress, and nitric oxide depletion, soluble guanylate cyclase (sGC) is a promising DKD drug target. sGC expression is specifically enriched in PT and stroma. In ZSF1 rats, pharmacological sGC activation confers considerable benefits over stimulation and is mechanistically related to improved oxidative stress regulation, resulting in enhanced downstream cGMP effects. Finally, we define sGC gene co-expression modules, which allow stratification of human kidney samples by DKD prevalence and disease-relevant measures such as kidney function, proteinuria, and fibrosis, underscoring the relevance of the sGC pathway to patients.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Humanos , Ratas , Animales , Guanilil Ciclasa Soluble/metabolismo , Guanilil Ciclasa Soluble/farmacología , Guanilil Ciclasa Soluble/uso terapéutico , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/patología , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Guanilato Ciclasa/farmacología , Riñón/metabolismo , Fibrosis
6.
Nat Commun ; 13(1): 4018, 2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35821371

RESUMEN

The kidney has tremendous capacity to repair after acute injury, however, pathways guiding adaptive and fibrotic repair are poorly understood. We developed a model of adaptive and fibrotic kidney regeneration by titrating ischemic injury dose. We performed detailed biochemical and histological analysis and profiled transcriptomic changes at bulk and single-cell level (> 110,000 cells) over time. Our analysis highlights kidney proximal tubule cells as key susceptible cells to injury. Adaptive proximal tubule repair correlated with fatty acid oxidation and oxidative phosphorylation. We identify a specific maladaptive/profibrotic proximal tubule cluster after long ischemia, which expresses proinflammatory and profibrotic cytokines and myeloid cell chemotactic factors. Druggability analysis highlights pyroptosis/ferroptosis as vulnerable pathways in these profibrotic cells. Pharmacological targeting of pyroptosis/ferroptosis in vivo pushed cells towards adaptive repair and ameliorates fibrosis. In summary, our single-cell analysis defines key differences in adaptive and fibrotic repair and identifies druggable pathways for pharmacological intervention to prevent kidney fibrosis.


Asunto(s)
Lesión Renal Aguda , Riñón , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Fibrosis , Humanos , Riñón/metabolismo , Regeneración , Análisis de la Célula Individual
7.
Nat Immunol ; 23(6): 947-959, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35552540

RESUMEN

Inflammation is an important component of fibrosis but immune processes that orchestrate kidney fibrosis are not well understood. Here we apply single-cell sequencing to a mouse model of kidney fibrosis. We identify a subset of kidney tubule cells with a profibrotic-inflammatory phenotype characterized by the expression of cytokines and chemokines associated with immune cell recruitment. Receptor-ligand interaction analysis and experimental validation indicate that CXCL1 secreted by profibrotic tubules recruits CXCR2+ basophils. In mice, these basophils are an important source of interleukin-6 and recruitment of the TH17 subset of helper T cells. Genetic deletion or antibody-based depletion of basophils results in reduced renal fibrosis. Human kidney single-cell, bulk gene expression and immunostaining validate a function for basophils in patients with kidney fibrosis. Collectively, these studies identify basophils as contributors to the development of renal fibrosis and suggest that targeting these cells might be a useful clinical strategy to manage chronic kidney disease.


Asunto(s)
Basófilos , Insuficiencia Renal Crónica , Animales , Fibrosis , Humanos , Riñón/metabolismo , Túbulos Renales , Ratones , Insuficiencia Renal Crónica/metabolismo , Análisis de la Célula Individual
8.
Annu Rev Physiol ; 84: 507-531, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-34843404

RESUMEN

The kidney maintains electrolyte, water, and acid-base balance, eliminates foreign and waste compounds, regulates blood pressure, and secretes hormones. There are at least 16 different highly specialized epithelial cell types in the mammalian kidney. The number of specialized endothelial cells, immune cells, and interstitial cell types might even be larger. The concerted interplay between different cell types is critical for kidney function. Traditionally, cells were defined by their function or microscopical morphological appearance. With the advent of new single-cell modalities such as transcriptomics, epigenetics, metabolomics, and proteomics we are entering into a new era of cell type definition. This new technological revolution provides new opportunities to classify cells in the kidney and understand their functions.


Asunto(s)
Células Endoteliales , Riñón , Animales , Presión Sanguínea , Células Epiteliales , Humanos , Riñón/fisiología , Mamíferos
9.
Ann N Y Acad Sci ; 1506(1): 74-97, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34605044

RESUMEN

Single cell biology has the potential to elucidate many critical biological processes and diseases, from development and regeneration to cancer. Single cell analyses are uncovering the molecular diversity of cells, revealing a clearer picture of the variation among and between different cell types. New techniques are beginning to unravel how differences in cell state-transcriptional, epigenetic, and other characteristics-can lead to different cell fates among genetically identical cells, which underlies complex processes such as embryonic development, drug resistance, response to injury, and cellular reprogramming. Single cell technologies also pose significant challenges relating to processing and analyzing vast amounts of data collected. To realize the potential of single cell technologies, new computational approaches are needed. On March 17-19, 2021, experts in single cell biology met virtually for the Keystone eSymposium "Single Cell Biology" to discuss advances both in single cell applications and technologies.


Asunto(s)
Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Congresos como Asunto/tendencias , Desarrollo Embrionario/fisiología , Informe de Investigación , Análisis de la Célula Individual/tendencias , Animales , Linaje de la Célula/fisiología , Humanos , Macrófagos/fisiología , Análisis de la Célula Individual/métodos
10.
Kidney Int ; 100(6): 1214-1226, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34534550

RESUMEN

A multitude of disease and therapy related factors drive the frequent development of kidney disorders in cancer patients. Along with chemotherapy, the newer targeted therapeutics can also cause kidney dysfunction through on and off-target mechanisms. Interestingly, among the small molecule inhibitors approved for the treatment of cancers that harbor BRAF-kinase activating mutations, vemurafenib can trigger tubular damage and acute kidney injury. BRAF is a proto-oncogene involved in cell growth. To investigate the underlying mechanisms, we developed cell culture and mouse models of vemurafenib kidney toxicity. At clinically relevant concentrations vemurafenib induces cell-death in transformed and primary mouse and human kidney tubular epithelial cells. In mice, two weeks of daily vemurafenib treatment causes moderate acute kidney injury with histopathological characteristics of kidney tubular epithelial cells injury. Importantly, kidney tubular epithelial cell-specific BRAF gene deletion did not influence kidney function under normal conditions or alter the severity of vemurafenib-associated kidney impairment. Instead, we found that inhibition of ferrochelatase, an enzyme involved in heme biosynthesis contributes to vemurafenib kidney toxicity. Ferrochelatase overexpression protected kidney tubular epithelial cells and conversely ferrochelatase knockdown increased the sensitivity to vemurafenib-induced kidney toxicity. Thus, our studies suggest that vemurafenib-associated kidney tubular epithelial cell dysfunction and kidney toxicity is BRAF-independent and caused, in part, by off-target ferrochelatase inhibition.


Asunto(s)
Ferroquelatasa , Proteínas Proto-Oncogénicas B-raf , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Indoles/toxicidad , Riñón/metabolismo , Ratones , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Sulfonamidas/toxicidad , Vemurafenib
11.
Nat Commun ; 12(1): 2277, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859189

RESUMEN

Determining the epigenetic program that generates unique cell types in the kidney is critical for understanding cell-type heterogeneity during tissue homeostasis and injury response. Here, we profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution. We show critical reliance of gene expression on distal regulatory elements (enhancers). We reveal key cell type-specific transcription factors and major gene-regulatory circuits for kidney cells. Dynamic chromatin and expression changes during nephron progenitor differentiation demonstrates that podocyte commitment occurs early and is associated with sustained Foxl1 expression. Renal tubule cells follow a more complex differentiation, where Hfn4a is associated with proximal and Tfap2b with distal fate. Mapping single nucleotide variants associated with human kidney disease implicates critical cell types, developmental stages, genes, and regulatory mechanisms. The single cell multi-omics atlas reveals key chromatin remodeling events and gene expression dynamics associated with kidney development.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Nefronas/crecimiento & desarrollo , Organogénesis/genética , Insuficiencia Renal Crónica/genética , Animales , Comunicación Celular , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Epigenómica , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Sitios Genéticos/genética , Estudio de Asociación del Genoma Completo , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Ratones , Nefronas/citología , Podocitos/fisiología , Polimorfismo de Nucleótido Simple , RNA-Seq , Insuficiencia Renal Crónica/patología , Análisis de la Célula Individual , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo
12.
J Am Soc Nephrol ; 32(6): 1279-1292, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33722930

RESUMEN

Over the last 5 years, single cell methods have enabled the monitoring of gene and protein expression, genetic, and epigenetic changes in thousands of individual cells in a single experiment. With the improved measurement and the decreasing cost of the reactions and sequencing, the size of these datasets is increasing rapidly. The critical bottleneck remains the analysis of the wealth of information generated by single cell experiments. In this review, we give a simplified overview of the analysis pipelines, as they are typically used in the field today. We aim to enable researchers starting out in single cell analysis to gain an overview of challenges and the most commonly used analytical tools. In addition, we hope to empower others to gain an understanding of how typical readouts from single cell datasets are presented in the published literature.


Asunto(s)
Análisis de Datos , Análisis de Secuencia de ARN , Análisis de la Célula Individual/métodos , Programas Informáticos , Visualización de Datos , Conjuntos de Datos como Asunto/normas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genómica , Humanos , Análisis de Componente Principal , Control de Calidad
13.
Eur J Immunol ; 51(2): 354-367, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32926407

RESUMEN

Peritoneal dialysis (PD) employs hypertonic glucose to remove excess water and uremic waste. Peritoneal membrane failure limits its long-term use. T-cell cytokines promote this decline. T-cell differentiation is critically determined by the microenvironment. We here study how PD-range hypertonic glucose regulates T-cell polarization and IL-17 production. In the human peritoneal cavity, CD3+ cell numbers increased in PD. Single cell RNA sequencing detected expression of T helper (Th) 17 signature genes RORC and IL23R. In vitro, PD-range glucose stimulated spontaneous and amplified cytokine-induced Th17 polarization. Osmotic controls l-glucose and d-mannose demonstrate that induction of IL-17A is a substance-independent, tonicity dose-dependent process. PD-range glucose upregulated glycolysis and increased the proportion of dysfunctional mitochondria. Blockade of reactive-oxygen species (ROS) prevented IL-17A induction in response to PD-range glucose. Peritoneal mesothelium cultured with IL-17A or IL17F produced pro-inflammatory cytokines IL-6, CCL2, and CX3CL1. In PD patients, peritoneal IL-17A positively correlated with CX3CL1 concentrations. PD-range glucose-stimulated, but neither identically treated Il17a-/- Il17f-/- nor T cells cultured with the ROS scavenger N-acetylcysteine enhanced mesothelial CX3CL1 expression. Our data delineate PD-range hypertonic glucose as a novel inducer of Th17 polarization in a mitochondrial-ROS-dependent manner. Modulation of tonicity-mediated effects of PD solutions may improve membrane survival.


Asunto(s)
Epitelio/inmunología , Glucosa/inmunología , Inflamación/inmunología , Interleucina-17/inmunología , Peritoneo/inmunología , Células Th17/inmunología , Animales , Células Cultivadas , Quimiocina CCL2/inmunología , Quimiocina CXCL1/inmunología , Femenino , Humanos , Interleucina-6/inmunología , Masculino , Manosa/inmunología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/inmunología , Diálisis Peritoneal/métodos , Especies Reactivas de Oxígeno/inmunología , Regulación hacia Arriba/inmunología
14.
Transpl Immunol ; 65: 101350, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33127498

RESUMEN

We previously induced long-term allograft acceptance in an allogeneic lung transplantation (LTx) model in miniature swine using perioperative non-myeloablative irradiation (IRR) combined with infusion of donor specific alloantigen. In order to improve clinical applicability, we delayed induction with irradiation in this study. Left sided single LTx was performed in minipigs. Group 1 received non-myeloablative irradiation (7Gy thymus and 1.5Gy whole body IRR) before LTx and a perioperative donor specific splenocyte infusion (SpTx). Group 2 received perioperative SpTx but delayed IRR three days after LTx. Group 3 was exposed to delayed IRR without SpTx. Whereas 4 out of 7 animals from the non-delayed group never rejected their grafts and were electively sacrificed on postoperative day (POD) +500, all animals from group 2 rejected their grafts before POD 108. In group 3, 3 out of 8 animals developed long-term allograft acceptance. In all groups, donor leukocyte chimerism peaked up to 20% in peripheral blood one hour after reperfusion of the lung. Group 1 maintained prolonged chimerism beyond POD 7, whereas chimerism levels in groups 2 and 3 decreased continuously thereafter. Delayed irradiation has the potential to improve long-term graft survival, yet not as efficient as a perioperative conditioning protocol.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Trasplante de Pulmón , Aloinjertos , Animales , Supervivencia de Injerto , Tolerancia Inmunológica , Porcinos , Porcinos Enanos , Quimera por Trasplante , Acondicionamiento Pretrasplante
15.
Cell Metab ; 33(2): 379-394.e8, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33301705

RESUMEN

Kidney disease is poorly understood because of the organ's cellular diversity. We used single-cell RNA sequencing not only in resolving differences in injured kidney tissue cellular composition but also in cell-type-specific gene expression in mouse models of kidney disease. This analysis highlighted major changes in cellular diversity in kidney disease, which markedly impacted whole-kidney transcriptomics outputs. Cell-type-specific differential expression analysis identified proximal tubule (PT) cells as the key vulnerable cell type. Through unbiased cell trajectory analyses, we show that PT cell differentiation is altered in kidney disease. Metabolism (fatty acid oxidation and oxidative phosphorylation) in PT cells showed the strongest and most reproducible association with PT cell differentiation and disease. Coupling of cell differentiation and the metabolism was established by nuclear receptors (estrogen-related receptor alpha [ESRRA] and peroxisomal proliferation-activated receptor alpha [PPARA]) that directly control metabolic and PT-cell-specific gene expression in mice and patient samples while protecting from kidney disease in the mouse model.


Asunto(s)
Enfermedades Renales/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Estrógenos/deficiencia , Receptor Relacionado con Estrógeno ERRalfa
16.
Biomolecules ; 10(11)2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33228017

RESUMEN

Peritoneal dialysis (PD) is limited by glucose-mediated peritoneal membrane (PM) fibrosis, angiogenesis, and ultrafiltration failure. Influencing PM integrity by pharmacologically targeting sodium-dependent glucose transporter (SGLT)-mediated glucose uptake has not been studied. In this study, wildtype C57Bl/6N mice were treated with high-glucose dialysate via an intraperitoneal catheter, with or without addition of selective SGLT2 inhibitor dapagliflozin. PM structural changes, ultrafiltration capacity, and peritoneal equilibration testing (PET) status for glucose, urea, and creatinine were analyzed. Expression of SGLT and facilitative glucose transporters (GLUT) was analyzed by real-time PCR, immunofluorescence, and immunohistochemistry. Peritoneal effluents were analyzed for cellular and cytokine composition. We found that peritoneal SGLT2 was expressed in mesothelial cells and in skeletal muscle. Dapagliflozin significantly reduced effluent transforming growth factor (TGF-ß) concentrations, peritoneal thickening, and fibrosis, as well as microvessel density, resulting in improved ultrafiltration, despite the fact that it did not affect development of high-glucose transporter status. In vitro, dapagliflozin reduced monocyte chemoattractant protein-1 release under high-glucose conditions in human and murine peritoneal mesothelial cells. Proinflammatory cytokine release in macrophages was reduced only when cultured in high-glucose conditions with an additional inflammatory stimulus. In summary, dapagliflozin improved structural and functional peritoneal health in the context of high-glucose PD.


Asunto(s)
Compuestos de Bencidrilo/uso terapéutico , Soluciones para Diálisis/toxicidad , Glucosa/toxicidad , Glucósidos/uso terapéutico , Fibrosis Peritoneal/tratamiento farmacológico , Peritoneo/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Transportador 2 de Sodio-Glucosa/biosíntesis , Adolescente , Animales , Compuestos de Bencidrilo/farmacología , Línea Celular Transformada , Células Cultivadas , Femenino , Glucósidos/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/metabolismo , Peritoneo/patología , Células RAW 264.7 , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Ultrafiltración/métodos
17.
Cell Signal ; 75: 109778, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32926960

RESUMEN

Peritoneal dialysis (PD) is a renal replacement therapy for patients with end-stage renal disease that is equivalent to hemodialysis with respect to adequacy, mortality, and other outcome parameters, yet providing superior quality-of-life measures and cost savings. However, long-term usage of the patient's peritoneal membrane as a dialyzer filter is unphysiological and leads to peritoneal fibrosis, which is a major factor of patient morbidity and PD technique failure, resulting in a transfer to hemodialysis or death. Peritoneal fibrosis pathophysiology involves chronic inflammation and the fibrotic process itself. Frequently, inflammation precedes membrane fibrosis development, although a bidirectional relationship of one inducing the other exists. This review aims at highlighting the histopathological definition of peritoneal fibrosis, outlining the interplay of fibrosis, angiogenesis and epithelial-to-mesenchymal transition (EMT), delineating important fibrogenic pathways involving Smad-dependent and Smad-independent transforming growth factor-ß (TGF-ß) as well as connective tissue growth factor (CTGF) signaling, and summarizing historic and recent studies of inflammatory pathways involving NOD-like receptor protein 3 (NLRP3)/interleukin (IL)-1ß, IL-6, IL-17, and other cytokines.


Asunto(s)
Inflamación/metabolismo , Fallo Renal Crónico , Neovascularización Patológica , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal , Animales , Citocinas/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/metabolismo , Transducción de Señal
18.
Sci Signal ; 13(635)2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32518144

RESUMEN

In this issue of Science Signaling, Lovisa et al. demonstrate the contribution of endothelial-to-mesenchymal transition (EndMT) to kidney fibrosis development. Their studies reveal the interdependence of endothelial and epithelial metabolism in health and disease.


Asunto(s)
Transición Epitelial-Mesenquimal , Enfermedades Renales , Endotelio , Fibrosis , Humanos , Riñón/patología , Enfermedades Renales/patología
19.
Clin Res Cardiol ; 109(8): 1035-1047, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32002632

RESUMEN

BACKGROUND: Dialysis patients are at increased risk of HF. However, diagnostic utility of NT-proBNP as a biomarker is decreased in patients on dialysis. GDF-15 and cNEP are biomarkers of distinct mechanisms that may contribute to HF pathophysiology in such cohorts. The aim of this study was to determine whether growth differentiation factor-15 (GDF-15) and circulating neprilysin (cNEP) improve the diagnosis of congestive heart failure (HF) in patients on dialysis. METHODS AND RESULTS: We compared circulating concentrations of NT-proBNP, GDF-15, and cNEP along with cNEP activity in patients on chronic dialysis without (n = 80) and with HF (n = 73), as diagnosed by clinical parameters and post-dialysis echocardiography. We used correlation, linear and logistic regression as well as receiver operating characteristic (ROC) analyses. Compared to controls, patients with HF had higher median values of NT-proBNP (16,216 [interquartile range, IQR = 27739] vs. 2883 [5866] pg/mL, p < 0.001), GDF-15 (7512 [7084] vs. 6005 [4892] pg/mL, p = 0.014), but not cNEP (315 [107] vs. 318 [124] pg/mL, p = 0.818). Median cNEP activity was significantly lower in HF vs. controls (0.189 [0.223] vs. 0.257 [0.166] nmol/mL/min, p < 0.001). In ROC analyses, a multi-marker model combining clinical covariates, NT-proBNP, GDF-15, and cNEP activity demonstrated best discrimination of HF from controls (AUC = 0.902, 95% CI 0.857-0.947, p < 0.001 vs. base model AUC = 0.785). CONCLUSION: We present novel comparative data on physiologically distinct circulating biomarkers for HF in patients on dialysis. cNEP activity but not concentration and GDF-15 provided incremental diagnostic information over clinical covariates and NT-proBNP and may aid in diagnosing HF in dialysis patients.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/sangre , Insuficiencia Cardíaca/sangre , Péptido Natriurético Encefálico/sangre , Neprilisina/sangre , Fragmentos de Péptidos/sangre , Diálisis Renal/efectos adversos , Biomarcadores/sangre , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Humanos , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/terapia , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Precursores de Proteínas , Curva ROC , Estudios Retrospectivos , Volumen Sistólico/fisiología
20.
Transpl Int ; 33(4): 376-390, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31705694

RESUMEN

Among factors determining long-term kidney allograft outcome, pretransplant renal replacement therapy (RRT) is the most easily modifiable. Previous studies analysing RRT modality impact on patient and graft survival are conflicting. Studies on allograft function are scarce, lack sufficient size and follow-up. We retrospectively studied patient and allograft survival together with allograft function and its decline in 2277 allograft recipients during 2000-2014. Pretransplant RRT modality ≥60 days as grouped into "no RRT" (n = 136), "haemodialysis (HD)" (n = 1847), "peritoneal dialysis (PD)" (n = 159), and "HD + PD" (n = 135) was evaluated. Kaplan-Meier analysis demonstrated superior 5-/10-/15-year patient (93.0/81.8/73.1% vs. 86.2/71.6/49.8%), death-censored graft (90.8/85.4/71.5% vs. 84.4/75.2/63.2%), and 1-year rejection-free graft survival (73.8% vs. 63.8%) in PD versus HD patients. Adjusted Cox regression revealed 34.5% [1.5-56.5%] lower hazards of death, whereas death-censored graft loss was similar [HR = 0.707 (0.469-1.064)], and rejection was less frequent [HR = 0.700 (0.508-0.965)]. Allografts showed higher 1-/3-/5-year estimated glomerular filtration rate (eGFR) in "PD" versus "HD" groups. Living donation benefit for allograft function was most pronounced in groups "no RRT" and "PD". Functional allograft decline (eGFR slope) was lowest for "PD". Allograft recipients on pretransplant PD versus HD demonstrated superior all-cause patient and rejection-free graft survival along with better allograft function (eGFR).


Asunto(s)
Fallo Renal Crónico , Trasplante de Riñón , Aloinjertos , Estudios de Cohortes , Supervivencia de Injerto , Humanos , Riñón , Fallo Renal Crónico/cirugía , Diálisis Renal , Estudios Retrospectivos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...