Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 608(7924): 766-777, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35948637

RESUMEN

Myocardial infarction is a leading cause of death worldwide1. Although advances have been made in acute treatment, an incomplete understanding of remodelling processes has limited the effectiveness of therapies to reduce late-stage mortality2. Here we generate an integrative high-resolution map of human cardiac remodelling after myocardial infarction using single-cell gene expression, chromatin accessibility and spatial transcriptomic profiling of multiple physiological zones at distinct time points in myocardium from patients with myocardial infarction and controls. Multi-modal data integration enabled us to evaluate cardiac cell-type compositions at increased resolution, yielding insights into changes of the cardiac transcriptome and epigenome through the identification of distinct tissue structures of injury, repair and remodelling. We identified and validated disease-specific cardiac cell states of major cell types and analysed them in their spatial context, evaluating their dependency on other cell types. Our data elucidate the molecular principles of human myocardial tissue organization, recapitulating a gradual cardiomyocyte and myeloid continuum following ischaemic injury. In sum, our study provides an integrative molecular map of human myocardial infarction, represents an essential reference for the field and paves the way for advanced mechanistic and therapeutic studies of cardiac disease.


Asunto(s)
Remodelación Atrial , Ensamble y Desensamble de Cromatina , Perfilación de la Expresión Génica , Infarto del Miocardio , Análisis de la Célula Individual , Remodelación Ventricular , Remodelación Atrial/genética , Estudios de Casos y Controles , Cromatina/genética , Epigenoma , Humanos , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factores de Tiempo , Remodelación Ventricular/genética
2.
Nature ; 589(7841): 281-286, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33176333

RESUMEN

Kidney fibrosis is the hallmark of chronic kidney disease progression; however, at present no antifibrotic therapies exist1-3. The origin, functional heterogeneity and regulation of scar-forming cells that occur during human kidney fibrosis remain poorly understood1,2,4. Here, using single-cell RNA sequencing, we profiled the transcriptomes of cells from the proximal and non-proximal tubules of healthy and fibrotic human kidneys to map the entire human kidney. This analysis enabled us to map all matrix-producing cells at high resolution, and to identify distinct subpopulations of pericytes and fibroblasts as the main cellular sources of scar-forming myofibroblasts during human kidney fibrosis. We used genetic fate-tracing, time-course single-cell RNA sequencing and ATAC-seq (assay for transposase-accessible chromatin using sequencing) experiments in mice, and spatial transcriptomics in human kidney fibrosis, to shed light on the cellular origins and differentiation of human kidney myofibroblasts and their precursors at high resolution. Finally, we used this strategy to detect potential therapeutic targets, and identified NKD2 as a myofibroblast-specific target in human kidney fibrosis.


Asunto(s)
Linaje de la Célula , Fibrosis/patología , Túbulos Renales/patología , Miofibroblastos/patología , Insuficiencia Renal Crónica/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Estudios de Casos y Controles , Diferenciación Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Masculino , Mesodermo/citología , Mesodermo/patología , Ratones , Miofibroblastos/metabolismo , Pericitos/citología , Pericitos/patología , RNA-Seq , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Análisis de la Célula Individual , Transcriptoma
3.
Development ; 149(9)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35417019

RESUMEN

Nephrotic syndrome (NS) is characterized by severe proteinuria as a consequence of kidney glomerular injury due to podocyte damage. In vitro models mimicking in vivo podocyte characteristics are a prerequisite to resolve NS pathogenesis. The detailed characterization of organoid podocytes resulting from a hybrid culture protocol showed a podocyte population that resembles adult podocytes and was superior compared with 2D counterparts, based on single-cell RNA sequencing, super-resolution imaging and electron microscopy. In this study, these next-generation podocytes in kidney organoids enabled personalized idiopathic nephrotic syndrome modeling, as shown by activated slit diaphragm signaling and podocyte injury following protamine sulfate, puromycin aminonucleoside treatment and exposure to NS plasma containing pathogenic permeability factors. Organoids cultured from cells of a patient with heterozygous NPHS2 mutations showed poor NPHS2 expression and aberrant NPHS1 localization, which was reversible after genetic correction. Repaired organoids displayed increased VEGFA pathway activity and transcription factor activity known to be essential for podocyte physiology, as shown by RNA sequencing. This study shows that organoids are the preferred model of choice to study idiopathic and congenital podocytopathies.


Asunto(s)
Síndrome Nefrótico , Células Madre Pluripotentes , Podocitos , Femenino , Humanos , Riñón/metabolismo , Masculino , Síndrome Nefrótico/genética , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Organoides , Células Madre Pluripotentes/metabolismo , Podocitos/metabolismo , Podocitos/patología
4.
Mol Syst Biol ; 20(2): 57-74, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38177382

RESUMEN

Although clinical applications represent the next challenge in single-cell genomics and digital pathology, we still lack computational methods to analyze single-cell or pathomics data to find sample-level trajectories or clusters associated with diseases. This remains challenging as single-cell/pathomics data are multi-scale, i.e., a sample is represented by clusters of cells/structures, and samples cannot be easily compared with each other. Here we propose PatIent Level analysis with Optimal Transport (PILOT). PILOT uses optimal transport to compute the Wasserstein distance between two individual single-cell samples. This allows us to perform unsupervised analysis at the sample level and uncover trajectories or cellular clusters associated with disease progression. We evaluate PILOT and competing approaches in single-cell genomics or pathomics studies involving various human diseases with up to 600 samples/patients and millions of cells or tissue structures. Our results demonstrate that PILOT detects disease-associated samples from large and complex single-cell or pathomics data. Moreover, PILOT provides a statistical approach to find changes in cell populations, gene expression, and tissue structures related to the trajectories or clusters supporting interpretation of predictions.


Asunto(s)
Algoritmos , Genómica , Humanos , Análisis por Conglomerados , Genómica/métodos
5.
Curr Opin Nephrol Hypertens ; 33(4): 420-426, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38587103

RESUMEN

PURPOSE OF REVIEW: Kidney fibrosis is a key pathological aspect and outcome of chronic kidney disease (CKD). The advent of multiomic analyses using human kidney tissue, enabled by technological advances, marks a new chapter of discovery in fibrosis research of the kidney. This review highlights the rapid advancements of single-cell and spatial multiomic techniques that offer new avenues for exploring research questions related to human kidney fibrosis development. RECENT FINDINGS: We recently focused on understanding the origin and transition of myofibroblasts in kidney fibrosis using single-cell RNA sequencing (scRNA-seq) [1] . We analysed cells from healthy human kidneys and compared them to patient samples with CKD. We identified PDGFRα+/PDGFRß+ mesenchymal cells as the primary cellular source of extracellular matrix (ECM) in human kidney fibrosis. We found several commonly shared cell states of fibroblasts and myofibroblasts and provided insights into molecular regulators. Novel single-cell and spatial multiomics tools are now available to shed light on cell lineages, the plasticity of kidney cells and cell-cell communication in fibrosis. SUMMARY: As further single-cell and spatial multiomic approaches are being developed, opportunities to apply these methods to human kidney tissues expand similarly. Careful design and optimisation of the multiomic experiments are needed to answer questions related to cell lineages, plasticity and cell-cell communication in kidney fibrosis.


Asunto(s)
Fibrosis , Riñón , Miofibroblastos , Análisis de la Célula Individual , Humanos , Riñón/patología , Riñón/metabolismo , Miofibroblastos/patología , Miofibroblastos/metabolismo , Análisis de la Célula Individual/métodos , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/metabolismo , Animales , Matriz Extracelular/metabolismo , Matriz Extracelular/patología
6.
Artículo en Inglés | MEDLINE | ID: mdl-38037533

RESUMEN

BACKGROUND AND HYPOTHESIS: Glucocorticoids are the treatment of choice for proteinuric patients with minimal-change disease (MCD) and primary focal and segmental glomerulosclerosis (FSGS). Immunosuppressive as well as direct effects on podocytes are believed to mediate their actions. In this study, we analyzed the anti-proteinuric effects of inhibition of the glucocorticoid receptor (GR) in glomerular epithelial cells, including podocytes. METHODS: We employed genetic and pharmacological approaches to inhibit the GR. Genetically, we used Pax8-Cre/GRfl/fl mice to specifically inactivate the GR in kidney epithelial cells. Pharmacologically, we utilized a glucocorticoid antagonist called mifepristone. RESULTS: Genetic inactivation of GR, specifically in kidney epithelial cells, using Pax8-Cre/GRfl/fl mice, ameliorated proteinuria following protein overload. We further tested the effects of pharmacological GR inhibition in three models and species: the puromycin-aminonucleoside-induced nephrosis model in rats, the protein overload model in mice and the inducible transgenic NTR/MTZ zebrafish larvae with specific and reversible podocyte injury. In all three models, both pharmacological GR activation and inhibition consistently and significantly ameliorated proteinuria. Additionally, we translated our findings to humans, where three nephrotic adult patients with MCD or primary FSGS with contraindications or insufficient responses to corticosteroids, were treated with mifepristone. This treatment resulted in a clinically relevant reduction of proteinuria. CONCLUSIONS: Thus, across multiple species and proteinuria models, both genetic and pharmacological GR inhibition was at least as effective as pronounced GR activation. While, the mechanism remains perplexing, GR inhibition may be a novel and targeted therapeutic approach to treat glomerular proteinuria potentially bypassing adverse actions of steroids.

7.
Kidney Int ; 101(2): 338-348, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34774554

RESUMEN

Chronic kidney disease (CKD) is accompanied with extensive cardiovascular calcification, in part correlating with functional vitamin K deficiency. Here, we sought to determine causes for vitamin K deficiency beyond reduced dietary intake. Initially, vitamin K uptake and distribution into circulating lipoproteins after a single administration of vitamin K1 plus K2 (menaquinone 4 and menaquinone 7, respectively) was determined in patients on dialysis therapy and healthy individuals. The patients incorporated very little menaquinone 7 but more menaquinone 4 into high density lipoprotein (HDL) and low-density lipoprotein particles than did healthy individuals. In contrast to healthy persons, HDL particles from the patients could not be spiked with menaquinone 7 in vitro and HDL uptake was diminished in osteoblasts. A reduced carboxylation activity (low vitamin K activity) of uremic HDL particles spiked with menaquinone 7 vs. that of controls was confirmed in a bioassay using human primary vascular smooth muscle cells. Kidney menaquinone 4 tissue levels were reduced in 5/6-nephrectomized versus sham-operated C57BL/6 mice after four weeks of a vitamin K rich diet. From the analyzed enzymes involved in vitamin K metabolism, kidney HMG-CoA reductase protein was reduced in both rats and patients with CKD. In a trial on the efficacy and safety of atorvastatin in 1051 patients with type 2 diabetes receiving dialysis therapy, no pronounced vitamin K deficiency was noted. However, the highest levels of PIVKA-II (biomarker of subclinical vitamin K deficiency) were noted when a statin was combined with a proton pump inhibitor. Thus, profound disturbances in lipoprotein mediated vitamin K transport and metabolism in uremia suggest that menaquinone 7 supplementation to patients on dialysis therapy has reduced efficacy.


Asunto(s)
Insuficiencia Renal Crónica , Deficiencia de Vitamina K , Vitamina K/metabolismo , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratas , Insuficiencia Renal Crónica/metabolismo , Distribución Tisular , Vitamina K/uso terapéutico , Vitamina K 1/metabolismo , Vitamina K 1/uso terapéutico , Vitamina K 2/metabolismo , Vitamina K 2/uso terapéutico , Deficiencia de Vitamina K/complicaciones , Deficiencia de Vitamina K/metabolismo
8.
Mol Syst Biol ; 17(1): e9730, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33502086

RESUMEN

Multi-omics datasets can provide molecular insights beyond the sum of individual omics. Various tools have been recently developed to integrate such datasets, but there are limited strategies to systematically extract mechanistic hypotheses from them. Here, we present COSMOS (Causal Oriented Search of Multi-Omics Space), a method that integrates phosphoproteomics, transcriptomics, and metabolomics datasets. COSMOS combines extensive prior knowledge of signaling, metabolic, and gene regulatory networks with computational methods to estimate activities of transcription factors and kinases as well as network-level causal reasoning. COSMOS provides mechanistic hypotheses for experimental observations across multi-omics datasets. We applied COSMOS to a dataset comprising transcriptomics, phosphoproteomics, and metabolomics data from healthy and cancerous tissue from eleven clear cell renal cell carcinoma (ccRCC) patients. COSMOS was able to capture relevant crosstalks within and between multiple omics layers, such as known ccRCC drug targets. We expect that our freely available method will be broadly useful to extract mechanistic insights from multi-omics studies.


Asunto(s)
Carcinoma de Células Renales/genética , Biología Computacional/métodos , Redes Reguladoras de Genes , Neoplasias Renales/genética , Carcinoma de Células Renales/metabolismo , Estudios de Casos y Controles , Perfilación de la Expresión Génica , Humanos , Neoplasias Renales/metabolismo , Metabolómica , Fosfoproteínas
9.
Nephrol Dial Transplant ; 37(4): 628-637, 2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33332571

RESUMEN

The field of single-cell technologies, in particular single-cell genomics with transcriptomics and epigenomics, and most recently single-cell proteomics, is rapidly growing and holds promise to advance our understanding of organ homoeostasis and disease, and facilitate the identification of novel therapeutic targets and biomarkers. This review offers an introduction to these technologies. In addition, as the size and complexity of the data require sophisticated computational methods for analysis and interpretation, we will also provide an overview of these methods and summarize the single-cell literature specifically pertaining to the kidney.


Asunto(s)
Epigenómica , Genómica , Biología Computacional/métodos , Epigenómica/métodos , Genómica/métodos , Humanos , Riñón , Metabolómica/métodos , Proteómica/métodos , Transcriptoma
10.
Basic Res Cardiol ; 116(1): 57, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34647168

RESUMEN

The adrenal glands participate in cardiovascular (CV) physiology and the pathophysiology of CV diseases through their effects on sodium and water metabolism, vascular tone and cardiac function. In the present study, we identified a new adrenal compound controlling mesenchymal cell differentiation that regulates osteoblastic differentiation in the context of vascular calcification. This peptide was named the "calcification blocking factor" (CBF) due to its protective effect against vascular calcification and is released from chromogranin A via enzymatic cleavage by calpain 1 and kallikrein. CBF reduced the calcium content of cells and thoracic aortic rings under calcifying culture conditions, as well as in aortas from animals treated with vitamin D and nicotine (VDN animals). Furthermore, CBF prevented vascular smooth muscle cell (VSMC) transdifferentiation into osteoblast-like cells within the vascular wall via the sodium-dependent phosphate transporter PIT-1 and by inhibition of NF-κB activation and the subsequent BMP2/p-SMAD pathway. Pulse pressure, a marker of arterial stiffness, was significantly decreased in VDN animals treated with CBF. In line with our preclinical data, CBF concentration is significantly reduced in diseases characterized by increased calcification, as shown in patients with chronic kidney disease. In preparation for clinical translation, the active site of the native 19-AS long native CBF was identified as EGQEEEED. In conclusion, we have identified the new peptide CBF, which is secreted from the adrenal glands and might prevent vascular calcification by inhibition of osteogenic transdifferentiation. The anti-calcific effects of CBF and short active site may therefore promote the development of new tools for the prevention and/or treatment of vascular calcification.


Asunto(s)
Transdiferenciación Celular , Calcificación Vascular , Animales , Células Cultivadas , Cromogranina A , Humanos , Músculo Liso Vascular , Miocitos del Músculo Liso , Calcificación Vascular/prevención & control
11.
J Am Soc Nephrol ; 31(12): 2773-2792, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32938648

RESUMEN

BACKGROUND: The roles of asymptomatic hyperuricemia or uric acid (UA) crystals in CKD progression are unknown. Hypotheses to explain links between UA deposition and progression of CKD include that (1) asymptomatic hyperuricemia does not promote CKD progression unless UA crystallizes in the kidney; (2) UA crystal granulomas may form due to pre-existing CKD; and (3) proinflammatory granuloma-related M1-like macrophages may drive UA crystal-induced CKD progression. METHODS: MALDI-FTICR mass spectrometry, immunohistochemistry, 3D confocal microscopy, and flow cytometry were used to characterize a novel mouse model of hyperuricemia and chronic UA crystal nephropathy with granulomatous nephritis. Interventional studies probed the role of crystal-induced inflammation and macrophages in the pathology of progressive CKD. RESULTS: Asymptomatic hyperuricemia alone did not cause CKD or drive the progression of aristolochic acid I-induced CKD. Only hyperuricemia with UA crystalluria due to urinary acidification caused tubular obstruction, inflammation, and interstitial fibrosis. UA crystal granulomas surrounded by proinflammatory M1-like macrophages developed late in this process of chronic UA crystal nephropathy and contributed to the progression of pre-existing CKD. Suppressing M1-like macrophages with adenosine attenuated granulomatous nephritis and the progressive decline in GFR. In contrast, inhibiting the JAK/STAT inflammatory pathway with tofacitinib was not renoprotective. CONCLUSIONS: Asymptomatic hyperuricemia does not affect CKD progression unless UA crystallizes in the kidney. UA crystal granulomas develop late in chronic UA crystal nephropathy and contribute to CKD progression because UA crystals trigger M1-like macrophage-related interstitial inflammation and fibrosis. Targeting proinflammatory macrophages, but not JAK/STAT signaling, can attenuate granulomatous interstitial nephritis.


Asunto(s)
Hiperuricemia/complicaciones , Hiperuricemia/patología , Nefritis Intersticial/etiología , Nefritis Intersticial/patología , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología , Animales , Enfermedades Asintomáticas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Granuloma/etiología , Granuloma/metabolismo , Granuloma/patología , Hiperuricemia/metabolismo , Ratones , Nefritis Intersticial/sangre , Insuficiencia Renal Crónica/sangre
12.
Kidney Int ; 96(1): 80-93, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31029503

RESUMEN

Beside the classical flat parietal epithelial cells (PECs), we investigated proximal tubular epithelial-like cells, a neglected subgroup of PECs. These cells, termed cuboidal PECs, make up the most proximal part of the proximal tubule and may also line parts of Bowman's capsule. Additionally, a third intermediate PEC subgroup was identified at the junction between the flat and cuboidal PEC subgroups at the tubular orifice. The transgenic mouse line PEC-rtTA labeled all three PEC subgroups. Here we show that the inducible Pax8-rtTA mouse line specifically labeled only cuboidal and intermediate PECs, but not flat PECs. In aging Pax8-rtTA mice, cell fate mapping showed no evidence for significant transdifferentiation from flat PECs to cuboidal or intermediate PECs or vice versa. In murine glomerular disease models of crescentic glomerulonephritis, and focal segmental glomerulosclerosis (FSGS), intermediate PECs became more numerous. These intermediate PECs preferentially expressed activation markers CD44 and Ki-67, suggesting that this subgroup of PECs was activated more easily than the classical flat PECs. In mice with FSGS, cuboidal and intermediate PECs formed sclerotic lesions. In patients with FSGS, cells forming the tip lesions expressed markers of intermediate PECs. These novel PEC subgroups form sclerotic lesions and were more prone to cellular activation compared to the classical flat PECs in disease. Thus, colonization of Bowman's capsule by cuboidal PECs may predispose to lesion formation and chronic kidney disease. We propose that tip lesions originate from this novel subgroup of PECs in patients with FSGS.


Asunto(s)
Células Epiteliales/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Glomérulos Renales/patología , Túbulos Renales Proximales/citología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Cápsula Glomerular/citología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Factor de Transcripción PAX8/genética , Adulto Joven
13.
Kidney Int ; 95(5): 1103-1119, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30827511

RESUMEN

Platelet-derived growth factors (PDGF) have been implicated in kidney disease progression. We previously found that PDGF-C is upregulated at sites of renal fibrosis and that antagonism of PDGF-C reduces fibrosis in the unilateral ureteral obstruction model. We studied the role of PDGF-C in collagen 4A3-/- ("Alport") mice, a model of progressive renal fibrosis with greater relevance to human kidney disease. Alport mice were crossbred with PDGF-C-/- mice or administered a neutralizing PDGF-C antibody. Both PDGF-C deficiency and neutralization reduced serum creatinine and blood urea nitrogen levels and mitigated glomerular injury, renal fibrosis, and renal inflammation. Unexpectedly, systolic blood pressure was also reduced in both Alport and wild-type mice treated with a neutralizing PDGF-C antibody. Neutralization of PDGF-C reduced arterial wall thickness in the renal cortex of Alport mice. Aortic rings isolated from anti-PDGF-C-treated wildtype mice exhibited reduced tension and faster relaxation than those of untreated mice. In vitro, PDGF-C upregulated angiotensinogen in aortic tissue and in primary hepatocytes and induced nuclear factor κB (NFκB)/p65-binding to the angiotensinogen promoter in hepatocytes. Neutralization of PDGF-C suppressed transcript expression of angiotensinogen in Alport mice and angiotensin II receptor type 1 in Alport and wildtype mice. Finally, administration of neutralizing PDGF-C antibodies ameliorated angiotensin II-induced hypertension in healthy mice. Thus, in addition to its key role in mediating renal fibrosis, we identified PDGF-C as a mediator of hypertension via effects on renal vasculature and on the renin-angiotensin system. The contribution to both renal fibrosis and hypertension render PDGF-C an attractive target in progressive kidney disease.


Asunto(s)
Hipertensión/patología , Riñón/patología , Linfocinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Angiotensinógeno/metabolismo , Animales , Presión Sanguínea/genética , Células Cultivadas , Colágeno Tipo IV/genética , Modelos Animales de Enfermedad , Fibrosis , Hepatocitos , Humanos , Hipertensión/etiología , Hipertensión/genética , Linfocinas/antagonistas & inhibidores , Masculino , Ratones , Ratones Noqueados , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Cultivo Primario de Células , Regulación hacia Arriba , Uréter/cirugía
14.
Kidney Int ; 96(2): 505-516, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31155155

RESUMEN

Recent developments in optical tissue clearing have been difficult to apply for the morphometric analysis of organs with high cellular content and small functional structures, such as the kidney. Here, we establish combinations of genetic and immuno-labelling for single cell identification, tissue clearing and subsequent de-clarification for histoimmunopathology and transmission electron microscopy. Using advanced light microscopy and computational analyses, we investigated a murine model of crescentic nephritis, an inflammatory kidney disease typified by immune-mediated damage to glomeruli leading to the formation of hypercellular lesions and the rapid loss of kidney function induced by nephrotoxic serum. Results show a graded susceptibility of the glomeruli, significant podocyte loss and capillary injury. These effects are associated with activation of parietal epithelial cells and formation of glomerular lesions that may evolve and obstruct the kidney tubule, thereby explaining the loss of kidney function. Thus, our work provides new high-throughput endpoints for the analysis of complex tissues with single-cell resolution.


Asunto(s)
Glomerulonefritis/patología , Técnicas de Preparación Histocitológica/métodos , Imagenología Tridimensional , Podocitos/fisiología , Análisis de la Célula Individual/métodos , Animales , Capilares , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fluorescencia , Colorantes Fluorescentes/química , Genes Reporteros/genética , Glomerulonefritis/inmunología , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Podocitos/ultraestructura
16.
Nephrol Dial Transplant ; 33(9): 1514-1525, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29635428

RESUMEN

Background: Vascular endothelial growth factor A (VEGF) is an essential growth factor during glomerular development and postnatal homeostasis. VEGF is secreted in high amounts by podocytes into the primary urine, back-filtered across the glomerular capillary wall to act on endothelial cells. So far it has been assumed that VEGF back-filtration is driven at a constant rate exclusively by diffusion. Methods: In the present work, glomerular VEGF back-filtration was investigated in vivo using a novel extended model based on endothelial fenestrations as surrogate marker for local VEGF concentrations. Single nephron glomerular filtration rate (SNGFR) and/or local filtration flux were manipulated by partial renal mass ablation, tubular ablation, and in transgenic mouse models of systemic or podocytic VEGF overexpression or reduction. Results: Our study shows positive correlations between VEGF back-filtration and SNGFR as well as effective filtration rate under physiological conditions along individual glomerular capillaries in rodents and humans. Conclusion: Our results suggest that an additional force drives VEGF back-filtration, potentially regulated by SNGFR.


Asunto(s)
Capilares/fisiopatología , Tasa de Filtración Glomerular/fisiología , Glomérulos Renales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Modelos Animales de Enfermedad , Glomérulos Renales/fisiopatología , Ratones , Ratones Noqueados , Nefrectomía
17.
J Am Soc Nephrol ; 28(5): 1408-1420, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27895155

RESUMEN

For several decades, glucocorticoids have been used empirically to treat rapid progressive GN. It is commonly assumed that glucocorticoids act primarily by dampening the immune response, but the mechanisms remain incompletely understood. In this study, we inactivated the glucocorticoid receptor (GR) specifically in kidney epithelial cells using Pax8-Cre/GRfl/fl mice. Pax8-Cre/GRfl/fl mice did not exhibit an overt spontaneous phenotype. In mice treated with nephrotoxic serum to induce crescentic nephritis (rapidly progressive GN), this genetic inactivation of the GR in kidney epithelial cells exerted renal benefits, including inhibition of albuminuria and cellular crescent formation, similar to the renal benefits observed with high-dose prednisolone in control mice. However, genetic inactivation of the GR in kidney epithelial cells did not induce the immunosuppressive effects observed with prednisolone. In vitro, prednisolone and the pharmacologic GR antagonist mifepristone each acted directly on primary cultures of parietal epithelial cells, inhibiting cellular outgrowth and proliferation. In wild-type mice, pharmacologic treatment with the GR antagonist mifepristone also attenuated disease as effectively as high-dose prednisolone without the systemic immunosuppressive effects. Collectively, these data show that glucocorticoids act directly on activated glomerular parietal epithelial cells in crescentic nephritis. Furthermore, we identified a novel therapeutic approach in crescentic nephritis, that of glucocorticoid antagonism, which was at least as effective as high-dose prednisolone with potentially fewer adverse effects.


Asunto(s)
Glomerulonefritis/tratamiento farmacológico , Glucocorticoides/farmacología , Glucocorticoides/uso terapéutico , Animales , Epitelio , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/fisiopatología , Ratones , Prednisolona , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/fisiología
18.
Curr Opin Nephrol Hypertens ; 25(4): 372-7, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27191350

RESUMEN

PURPOSE OF REVIEW: Multiple studies have established the beneficial role of mesenchymal stem cell (MSC) therapy for kidney injury. In this review we will discuss the recent identification of Gli1 as a marker for perivascular MSC and the role of this cell population in kidney fibrosis. RECENT FINDINGS: Recent studies demonstrate that expression of the hedgehog transcriptional activator Gli1 specifically marks perivascular MSC. Genetic fate tracing of MSC in kidney injury revealed their contribution to the myofibroblast pool whereas ablation of MSC reduced kidney fibrosis. Furthermore, strong evidence suggests that pharmacologically targeting Gli proteins inhibits cell-cycle progression of myofibroblasts in kidney fibrosis and is a promising therapeutic strategy in chronic kidney disease. SUMMARY: Although there is tremendous excitement about MSC as cellular therapy in kidney injury it has been shown that resident perivascular MSC are a major source of myofibroblasts and a novel therapeutic target in kidney fibrosis. While resident kidney MSC might also be involved in capillary rarefaction after injury and during fibrosis progression their potential role in kidney repair, angiogenesis, and regeneration remains unclear. Further studies are needed to identify the molecular pathways that control the profibrotic versus proregenerative role of resident MSC in kidney injury and repair.


Asunto(s)
Lesión Renal Aguda/metabolismo , Células Madre Mesenquimatosas/metabolismo , Miofibroblastos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Fibrosis , Humanos , Riñón/metabolismo , Riñón/patología , Regeneración
19.
Kidney Int ; 88(5): 990-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25853334

RESUMEN

Parietal epithelial cells (PECs) are involved in the development of sclerotic lesions in primary focal and segmental glomerulosclerosis (FSGS). Here, the role of PECs was explored in the more common secondary FSGS lesions in 68 patient biopsies, diagnosed with 11 different frequently or rarely encountered glomerular pathologies and additional secondary FSGS lesions. For each biopsy, one section was quadruple stained for PECs (ANXA3), podocytes (synaptopodin), PEC matrix (LKIV69), and Hoechst (nuclei), and a second was quadruple stained for activated PECs (CD44 and cytokeratin-19), PEC matrix, and nuclei. In all lesions, cellular adhesions (synechiae) between Bowman's capsule and the tuft were formed by cells expressing podocyte and/or PEC markers. Cells expressing PEC markers were detected in all FSGS lesions independent of the underlying glomerular disease and often stained positive for markers of activation. Small FSGS lesions, which were hardly identified on PAS sections previously, were detectable by immunofluorescent staining using PEC markers, potentially improving the diagnostic sensitivity to identify these lesions. Thus, similar patterns of cells expressing podocyte and/or PEC markers were found in the formation of secondary FSGS lesions independent of the underlying glomerular disease. Hence, our findings support the hypothesis that FSGS lesions follow a final cellular pathway to nephron loss that includes involvement of cells expressing PEC markers.


Asunto(s)
Células Epiteliales/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Glomérulos Renales/patología , Adulto , Anciano , Anexina A3/análisis , Cápsula Glomerular/patología , Adhesión Celular , Núcleo Celular/patología , Claudina-1/análisis , Células Epiteliales/química , Femenino , Técnica del Anticuerpo Fluorescente , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Humanos , Receptores de Hialuranos/análisis , Queratina-19/análisis , Glomérulos Renales/química , Masculino , Proteínas de Microfilamentos/análisis , Persona de Mediana Edad , Podocitos/química , Podocitos/patología , Coloración y Etiquetado , Adulto Joven
20.
Curr Opin Nephrol Hypertens ; 24(3): 208-16, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25887902

RESUMEN

PURPOSE OF REVIEW: At first sight, the glomerular filter appears like a problem that should be easily solved. The majority of researchers view the filter like an impermeable wall perforated by specialized and size-selective pores (pore model). However, the fact that this model is in conflict with many of the experimental findings suggests that it may not yet be complete. RECENT FINDINGS: In the more recent electrokinetic model, we have proposed including electrical effects (streaming potentials). The present review investigates how this can provide a relatively simple mechanistic explanation for the great majority of the so far unexplained characteristics of the filter, for example why the filter never clogs. SUMMARY: Understanding how the glomerular filter functions is a prerequisite to investigate the pathogenesis of proteinuric glomerular diseases and the link between glomerular proteinuria and cardiovascular disease.


Asunto(s)
Barrera de Filtración Glomerular/fisiología , Tasa de Filtración Glomerular/fisiología , Enfermedades Renales/fisiopatología , Glomérulos Renales/fisiología , Proteinuria/metabolismo , Animales , Endotelio/metabolismo , Humanos , Enfermedades Renales/patología , Glomérulos Renales/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA