Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Am J Physiol Renal Physiol ; 326(3): F326-F337, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38205542

RESUMO

Acute kidney injury activates both proliferative and antiproliferative pathways, the consequences of which are not fully elucidated. If an initial proliferation of the renal epithelium is necessary for the successful repair, the persistence of proliferation markers is associated with the occurrence of chronic kidney disease. We hypothesized that proliferation in stress conditions impacts cell viability and renal outcomes. We found that proliferation is associated with cell death after various stresses in kidney cells. In vitro, the ATP/ADP ratio oscillates reproducibly throughout the cell cycle, and cell proliferation is associated with a decreased intracellular ATP/ADP ratio. In vivo, transcriptomic data from transplanted kidneys revealed that proliferation was strongly associated with a decrease in the expression of the mitochondria-encoded genes of the oxidative phosphorylation pathway, but not of the nucleus-encoded ones. These observations suggest that mitochondrial function is a limiting factor for energy production in proliferative kidney cells after injury. The association of increased proliferation and decreased mitochondrial function was indeed associated with poor renal outcomes. In summary, proliferation is an energy-demanding process impairing the cellular ability to cope with an injury, highlighting proliferative repair and metabolic recovery as indispensable and interdependent features for successful kidney repair.NEW & NOTEWORTHY ATP depletion is a hallmark of acute kidney injury. Proliferation is instrumental to kidney repair. We show that ATP levels vary during the cell cycle and that proliferation sensitizes renal epithelial cells to superimposed injuries in vitro. More proliferation and less energy production by the mitochondria are associated with adverse outcomes in injured kidney allografts. This suggests that controlling the timing of kidney repair might be beneficial to mitigate the extent of acute kidney injury.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Humanos , Rim/metabolismo , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Células Epiteliais/metabolismo , Proliferação de Células , Trifosfato de Adenosina/metabolismo , Traumatismo por Reperfusão/metabolismo
2.
Kidney Int ; 101(4): 720-732, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35090878

RESUMO

To guide the development of therapeutic interventions for acute kidney injury, elucidating the deleterious pathways of this global health problem is highly warranted. Emerging evidence has indicated a pivotal role of endothelial dysfunction in the etiology of this disease. We found that the class III semaphorin SEMA3C was ectopically upregulated with full length protein excreted into the blood and truncated protein secreted into the urine upon kidney injury and hypothesized a role for SEAM3C in acute kidney injury. Sema3c was genetically abrogated during acute kidney injury and subsequent kidney morphological and functional defects in two well-characterized models of acute kidney injury; warm ischemia/reperfusion and folic acid injection were analyzed. Employing a beta actin-dependent, inducible knockout of Sema3c, we demonstrate that in acute kidney injury SEMA3C promotes interstitial edema, leucocyte infiltration and tubular injury. Additionally, intravital microscopy combined with Evans Blue dye extravasation and primary culture of magnetically sorted peritubular endothelial cells identified a novel role for SEMA3C in promoting vascular permeability. Thus, our study points to microvascular permeability as an important driver of injury in acute kidney injury, and to SEMA3C as a novel permeability factor and potential target for therapeutic intervention.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Semaforinas , Injúria Renal Aguda/genética , Injúria Renal Aguda/prevenção & controle , Animais , Permeabilidade Capilar , Células Endoteliais/metabolismo , Feminino , Humanos , Rim/metabolismo , Masculino , Camundongos , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/prevenção & controle , Semaforinas/genética , Semaforinas/metabolismo
3.
J Am Soc Nephrol ; 31(1): 85-100, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31690575

RESUMO

BACKGROUND: The matricellular protein periostin has been associated with CKD progression in animal models and human biopsy specimens. Periostin functions by interacting with extracellular matrix components to drive collagen fibrillogenesis and remodeling or by signaling through cell-surface integrin receptors to promote cell adhesion, migration, and proliferation. However, its role in AKI is unknown. METHODS: We used mice with conditional tubule-specific overexpression of periostin or knockout mice lacking periostin expression in the renal ischemia-reperfusion injury model, and primary cultures of isolated tubular cells in a hypoxia-reoxygenation model. RESULTS: Tubular epithelial cells showed strong production of periostin during the repair phase of ischemia reperfusion. Periostin overexpression protected mice from renal injury compared with controls, whereas knockout mice showed increased tubular injury and deteriorated renal function. Periostin interacted with its receptor, integrin-ß1, to inhibit tubular cell cycle arrest and apoptosis in in vivo and in vitro models. After ischemia-reperfusion injury, periostin-overexpressing mice exhibited diminished expression of proinflammatory molecules and had more F4/80+ macrophages compared with knockout mice. Macrophages from periostin-overexpressing mice showed increased proliferation and expression of proregenerative factors after ischemia-reperfusion injury, whereas knockout mice exhibited the opposite. Coculturing a macrophage cell line with hypoxia-treated primary tubules overexpressing periostin, or treating such macrophages with recombinant periostin, directly induced macrophage proliferation and expression of proregenerative molecules. CONCLUSIONS: In contrast to the detrimental role of periostin in CKD, we discovered a protective role of periostin in AKI. Our findings suggest periostin may be a novel and important mediator of mechanisms controlling renal repair after AKI.


Assuntos
Injúria Renal Aguda , Moléculas de Adesão Celular/fisiologia , Proliferação de Células , Macrófagos/fisiologia , Injúria Renal Aguda/etiologia , Animais , Modelos Animais de Doenças , Rim/irrigação sanguínea , Masculino , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia
4.
J Cell Mol Med ; 23(11): 7279-7288, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31469511

RESUMO

Chronic allograft dysfunction (CAD), defined as the replacement of functional renal tissue by extracellular matrix proteins, remains the first cause of graft loss. The aim of our study was to explore the potential role of the cannabinoid receptor 1 (CB1) during CAD. We retrospectively quantified CB1 expression and correlated it with renal fibrosis in 26 kidney-transplanted patients who underwent serial routine kidney biopsies. Whereas CB1 expression was low in normal kidney grafts, it was highly expressed during CAD, especially in tubular cells. CB1 expression significantly increased early on after transplantation, from day 0 (D0) to month 3 post-transplant (M3) (22.5% ± 15.4% vs 33.4% ± 13.8%, P < .01), and it remained stable thereafter. CB1 expression correlated with renal fibrosis at M3 (P = .04). In an in vitro model of tacrolimus-mediated fibrogenesis by tubular cells, we found that tacrolimus treatment significantly induced mRNA and protein expression of CB1 concomitantly to col3a1 and col4a3 up regulation. Administration of rimonabant, a CB1 antagonist, blunted collagen synthesis by tubular cells (P < .05). Overall, our study strongly suggests an involvement of the cannabinoid system in the progression of fibrosis during CAD and indicates the therapeutic potential of CB1 antagonists in this pathology.


Assuntos
Fibrose/etiologia , Transplante de Rim/efeitos adversos , Disfunção Primária do Enxerto/complicações , Receptor CB1 de Canabinoide/metabolismo , Animais , Células Cultivadas , Doença Crônica , Feminino , Fibrose/metabolismo , Fibrose/patologia , Humanos , Imunossupressores/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Disfunção Primária do Enxerto/cirurgia , Receptor CB1 de Canabinoide/genética , Estudos Retrospectivos , Tacrolimo/toxicidade
5.
Cell Physiol Biochem ; 47(4): 1338-1351, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29929186

RESUMO

BACKGROUND/AIMS: Fatty acid oxidation (FAO), the main source of energy produced by tubular epithelial cells in the kidney, was found to be defective in tubulo-interstitial samples dissected out in kidney biopsies from patients with chronic kidney disease (CKD). Experimental data indicated that this decrease was a strong determinant of renal fibrogenesis, hence a focus for therapeutic interventions. Nevertheless, whether persistently differentiated renal tubules, surviving in a pro-fibrotic environment, also suffer from a decrease in FAO, is currently unknown. METHODS: To address this question, we isolated proximal tubules captured ex vivo on the basis of the expression of an intact brush border antigen (Prominin-1) in C57BL6/J mice subjected to a controlled, two-hit model of renal fibrosis (reversible ischemic acute kidney injury (AKI) or sham surgery, followed by angiotensin 2 administration). A transcriptomic high throughput sequencing was performed on total mRNA from these cells, and on whole kidneys. RESULTS: In contrast to mice subjected to sham surgery, mice with a history of AKI displayed histologically more renal fibrosis when exposed to angiotensin 2. High throughput RNA sequencing, principal component analysis and clustering showed marked consistency within experimental groups. As expected, FAO transcripts were decreased in whole fibrotic kidneys. Surprisingly, however, up- rather than down-regulation of metabolic pathways (oxidative phosphorylation, fatty acid metabolism, glycolysis, and PPAR signalling pathway) was a hallmark of the differentiated tubules captured from fibrotic kidneys. Immunofluorescence co-staining analysis confirmed that the expression of FAO enzymes was dependent of tubular trophicity. CONCLUSIONS: These data suggest that in differentiated proximal tubules energetic hyperactivity is promoted concurrently with organ fibrogenesis.


Assuntos
Injúria Renal Aguda/metabolismo , Ácidos Graxos/metabolismo , Túbulos Renais Proximais/metabolismo , Antígeno AC133/metabolismo , Injúria Renal Aguda/patologia , Animais , Sobrevivência Celular , Túbulos Renais Proximais/patologia , Camundongos , Oxirredução
6.
J Immunol ; 196(1): 168-81, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26608921

RESUMO

Calpains are intracellular proteases that play a key role in inflammation/immunity. Rare studies show that they are partially externalized. However, the mechanism of this secretion and the functions of exteriorized calpains remain poorly understood. In this study, we found that mouse and human lymphocytes secreted calpains through an ABCA1-driven process. In turn, extracellular calpains inhibited IL-17A expression. We were able to attribute this function to a cleavage of the TLR2 extracellular domain, which prevented TLR2-induced transcription of molecules essential for IL-17A induction. Calpain exteriorization and TLR2 cleavage were critical for the control of IL-17A expression by low doses of IL-2. By using newly developed transgenic mice in which extracellular calpains are specifically inactivated, we provide evidence for the relevance of calpain externalization in vivo in regulating IL-17A expression and function in experimental sterile peritonitis and autoimmune arthritis, respectively. Thus, this study identifies calpain exteriorization as a potential target for immune modulation.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/biossíntese , Calpaína/metabolismo , Interleucina-17/biossíntese , Linfócitos T/imunologia , Receptor 2 Toll-Like/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Animais , Artrite Experimental , Linhagem Celular , Proliferação de Células , Regulação da Expressão Gênica , Células HEK293 , Humanos , Inflamação/imunologia , Mediadores da Inflamação/imunologia , Interleucina-17/genética , Interleucina-2/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/imunologia , Interferência de RNA , RNA Interferente Pequeno , Baço/citologia
7.
J Am Soc Nephrol ; 28(12): 3563-3578, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28928136

RESUMO

FSGS, the most common primary glomerular disorder causing ESRD, is a complex disease that is only partially understood. Progressive sclerosis is a hallmark of FSGS, and genetic tracing studies have shown that parietal epithelial cells participate in the formation of sclerotic lesions. The loss of podocytes triggers a focal activation of parietal epithelial cells, which subsequently form cellular adhesions with the capillary tuft. However, in the absence of intrinsic podocyte alterations, the origin of the pathogenic signal that triggers parietal epithelial cell recruitment remains elusive. In this study, investigation of the role of the endothelial PAS domain-containing protein 1 (EPAS1), a regulatory α subunit of the hypoxia-inducible factor complex, during angiotensin II-induced hypertensive nephropathy provided novel insights into FSGS pathogenesis in the absence of a primary podocyte abnormality. We infused angiotensin II into endothelial-selective Epas1 knockout mice and their littermate controls. Although the groups presented with identical high BP, endothelial-specific Epas1 gene deletion accentuated albuminuria with severe podocyte lesions and recruitment of pathogenic parietal glomerular epithelial cells. These lesions and dysfunction of the glomerular filtration barrier were associated with FSGS in endothelial Epas1-deficient mice only. These results indicate that endothelial EPAS1 has a global protective role during glomerular hypertensive injuries without influencing the hypertensive effect of angiotensin II. Furthermore, these findings provide proof of principle that endothelial-derived signaling can trigger FSGS and illustrate the potential importance of the EPAS1 endothelial transcription factor in secondary FSGS.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Epiteliais/citologia , Regulação da Expressão Gênica , Glomerulosclerose Segmentar e Focal/metabolismo , Hipertensão/metabolismo , Glomérulos Renais/metabolismo , Albuminas/análise , Angiotensina II/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Pressão Sanguínea , Diferenciação Celular , Cruzamentos Genéticos , Progressão da Doença , Células Epiteliais/metabolismo , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Podócitos/metabolismo , Telemetria
8.
J Am Soc Nephrol ; 28(6): 1723-1728, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28082518

RESUMO

Vancomycin is a widely prescribed antibiotic, but the exact nature of vancomycin-associated nephrotoxicity is unclear, in particular when considering the frequent coadministration of aminoglycosides. We describe here the initial case of a 56-year-old woman with normal renal function developing unexplained ARF without hypovolemia after administration of vancomycin without coadministration of aminoglycosides. Studying the patient's renal biopsy specimen, we ascertained that obstructive tubular casts composed of noncrystal nanospheric vancomycin aggregates entangled with uromodulin explained the vancomycin-associated ARF. We developed in parallel a new immunohistologic staining technique to detect vancomycin in renal tissue and confirmed retrospectively that deleterious vancomycin-associated casts existed in eight additional patients with acute tubular necrosis in the absence of hypovolemia. Concomitant high vancomycin trough plasma levels had been observed in each patient. We also reproduced experimentally the toxic and obstructive nature of vancomycin-associated cast nephropathy in mice, which we detected using different in vivo imaging techniques. In conclusion, the interaction of uromodulin with nanospheric vancomycin aggregates represents a new mode of tubular cast formation, revealing the hitherto unsuspected mechanism of vancomycin-associated renal injury.


Assuntos
Antibacterianos/efeitos adversos , Nefropatias/induzido quimicamente , Vancomicina/efeitos adversos , Antibacterianos/metabolismo , Feminino , Humanos , Nefropatias/patologia , Pessoa de Meia-Idade , Uromodulina/metabolismo , Vancomicina/metabolismo
9.
J Am Soc Nephrol ; 28(2): 545-556, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27451286

RESUMO

Acute tubular damage is a major cause of renal failure, especially at the early phase of kidney transplant when ischemia-reperfusion injury and cyclosporin A toxicity may coexist. The mechanisms of the latter are largely unknown. Using an mRNA microarray on microdissected tubules from a rat model of cyclosporin A toxicity to describe the related epithelial-specific transcriptional signature in vivo, we found that cyclosporin A induces pathways dependent on the transcription factor ATF4 and identified nuclear protein transcriptional regulator 1 (Nupr1), a stress response gene induced by ATF4, as the gene most strongly upregulated. Upon cyclosporin A treatment, Nupr1-deficient mice exhibited worse renal tubular lesions than wild-type mice. In primary cultures treated with cyclosporin A, renal tubular cells isolated from Nupr1-deficient mice exhibited more apoptosis and ATP depletion than cells from wild-type mice. Furthermore, cyclosporin A decreased protein synthesis and abolished proliferation in wild-type tubular cells, but only reduced proliferation in Nupr1-deficient cells. Compared with controls, mouse models of ischemia-reperfusion injury, urinary obstruction, and hypertension exhibited upregulated expression of renal NUPR1, and cyclosporin A induced Nupr1 expression in cultured human tubular epithelial cells. Finally, immunohistochemical analysis revealed strong expression of NUPR1 in the nuclei of renal proximal tubules of injured human kidney allografts, but not in those of stable allografts. Taken together, these results suggest that epithelial expression of NUPR1 has a protective role in response to injury after renal transplant and, presumably, in other forms of acute tubular damage.


Assuntos
Ciclosporina/toxicidade , Proteínas de Ligação a DNA/genética , Nefropatias/induzido quimicamente , Nefropatias/genética , Proteínas de Neoplasias/genética , Animais , Humanos , Camundongos , Estresse Fisiológico
10.
Kidney Int ; 91(5): 1146-1158, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28111009

RESUMO

Crescentic glomerulonephritis is a life-threatening renal disease that has been extensively studied by the experimental anti-glomerular basement membrane glomerulonephritis (anti-GBM-GN) model. Although T cells have a significant role in this model, athymic/nude mice and rats still develop severe renal disease. Here we further explored the contribution of intrinsic renal cells in the development of T-cell-independent GN lesions. Anti-GBM-GN was induced in three strains of immune-deficient mice (Rag2-/-, Rag2-/-Il2rg-/-, and Rag2-/-Il2rb-/-) that are devoid of either T/B cells or T/B/NK cells. The Rag2-/-Il2rg-/- or Rag2-/-Il2rb-/- mice harbor an additional deletion of either the common gamma chain (γC) or the interleukin-2 receptor ß subunit (IL-2Rß), respectively, impairing IL-15 signaling in particular. As expected, all these strains developed severe anti-GBM-GN. Additionally, bone marrow replenishment experiments allowed us to deduce a protective role for the glomerular-expressed γC during anti-GBM-GN. Given that IL-15 has been found highly expressed in nephritic kidneys despite the absence of lymphocytes, we then studied this cytokine in vitro on primary cultured podocytes from immune-deficient mice (Rag2-/-Il2rg-/- and Rag2-/-Il2rb-/-) compared to controls. IL-15 induced downstream activation of JAK1/3 and SYK in primary cultured podocytes. IL-15-dependent JAK/SYK induction was impaired in the absence of γC or IL-2Rß. We found γC largely induced on podocytes during human glomerulonephritis. Thus, renal lesions are indeed modulated by intrinsic glomerular cells through the γC/IL-2Rß receptor response, to date classically described only in immune cells.


Assuntos
Proteínas de Ligação a DNA/imunologia , Glomerulonefrite/imunologia , Subunidade gama Comum de Receptores de Interleucina/imunologia , Subunidade beta de Receptor de Interleucina-2/imunologia , Glomérulos Renais/imunologia , Podócitos/imunologia , Animais , Autoanticorpos/toxicidade , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Imunofluorescência , Glomerulonefrite/induzido quimicamente , Glomerulonefrite/metabolismo , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Interleucina-15/imunologia , Interleucina-15/metabolismo , Subunidade beta de Receptor de Interleucina-2/genética , Janus Quinase 1/metabolismo , Janus Quinase 3/metabolismo , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Células Matadoras Naturais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Podócitos/metabolismo , Cultura Primária de Células , Transdução de Sinais , Quinase Syk/metabolismo
11.
Ren Fail ; 38(4): 521-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26905096

RESUMO

It has been suggested that bone marrow derived stem cells have the ability to engraft the kidney and improve the outcome of severe acute kidney injury (AKI) in mice exposed to high doses of cisplatin, providing hope for cancer patients in whom irreversible renal damage occasionally occurs following the use of this highly effective anti-tumor drug. We tested the therapeutic potential of bone marrow derived cells injected during the acute phase (day 3 after cisplatin administration) of experimentally-induced AKI in C57Bl6/J mice, characterized by massive tubular necrosis, apoptosis, and a low proliferation capacity. We failed to show any benefit of bone marrow derived cells versus a regular homogenate of intact renal cells, or normal saline. Using cell tracers and flow cytometry, we demonstrated that bone marrow derived cells did indeed home to the bone marrow of the recipients but failed to settle in the kidney. Conversely, renal cells homed to injured kidneys. However, neither cell therapy protected the animals against cisplatin-induced death. We therefore question the short-term efficacy of bone marrow derived cells used to repair established injuries of the tubular epithelium.


Assuntos
Injúria Renal Aguda/cirurgia , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Túbulos Renais/fisiologia , Regeneração , Injúria Renal Aguda/induzido quimicamente , Animais , Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Feminino , Rim/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
12.
BMC Cell Biol ; 16: 12, 2015 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-25881040

RESUMO

BACKGROUND: Experimental models are inevitably a compromise between accurately reproducing a pathological situation and schematically simplifying it, which is intended to provide both relevance and conclusiveness. In-vivo models are very relevant, but multiple cell-types undergoing various changes may hinder the observation of individual molecular events. RESULTS: Here, we describe a method for analyzing and isolating specific cell types from the kidney and studying the phenotype they have acquired in vivo. Using flow cytometry, immunofluorescence, and RT-PCR, we show that our method is suitable for studying and isolating proximal tubular cells with an anti Prominin-1 antibody. Kidneys are subjected to mechanical dissociation followed by flow-cytometry analysis. Hundreds of thousands of proximal tubular cells are then isolated by magnetic separation followed by direct analysis or primary cell culture. Using our method, we detect phenotypic changes in the proximal tubular cells after renal ischemia reperfusion, and we isolate the proximal tubular cells, with a purity over 80%. CONCLUSIONS: This method is efficient, quick, simple, and cheap, and should be useful for studying cell-type specific parameters after in vivo experimental studies. It is also a simple method to obtain a specific primary cell culture from any animal strain.


Assuntos
Túbulos Renais Proximais/citologia , Antígeno AC133 , Animais , Anticorpos/imunologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Separação Celular , Células Cultivadas , Citometria de Fluxo , Imunofluorescência , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Receptores de Hialuronatos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/imunologia , Peptídeos/metabolismo , Fenótipo
13.
Kidney Int ; 88(1): 72-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25760323

RESUMO

Chronic kidney disease, secondary to renal fibrogenesis, is a burden on public health. There is a need to explore new therapeutic pathways to reduce renal fibrogenesis. To study this, we used unilateral ureteral obstruction (UUO) in mice as an experimental model of renal fibrosis and microarray analysis to compare gene expression in fibrotic and normal kidneys. The cannabinoid receptor 1 (CB1) was among the most upregulated genes in mice, and the main endogenous CB1 ligand (2-arachidonoylglycerol) was significantly increased in the fibrotic kidney. Interestingly, CB1 expression was highly increased in kidney biopsies of patients with IgA nephropathy, diabetes, and acute interstitial nephritis. Both genetic and pharmacological knockout of CB1 induced a profound reduction in renal fibrosis during UUO. While CB2 is also involved in renal fibrogenesis, it did not potentiate the role of CB1. CB1 expression was significantly increased in myofibroblasts, the main effector cells in renal fibrogenesis, upon TGF-ß1 stimulation. The decrease in renal fibrosis during CB1 blockade could be explained by a direct action on myofibroblasts. CB1 blockade reduced collagen expression in vitro. Rimonabant, a selective CB1 endocannabinoid receptor antagonist, modulated the macrophage infiltrate responsible for renal fibrosis in UUO through a decrease in monocyte chemoattractant protein-1 synthesis. Thus, CB1 has a major role in the activation of myofibroblasts and may be a new target for treating chronic kidney disease.


Assuntos
Fibrose/genética , Rim/patologia , Miofibroblastos/metabolismo , RNA Mensageiro/metabolismo , Receptor CB1 de Canabinoide/genética , Doença Aguda , Animais , Ácidos Araquidônicos , Células Cultivadas , Quimiocina CCL2/metabolismo , Colágeno/metabolismo , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Endocanabinoides , Fibrose/metabolismo , Fibrose/patologia , Perfilação da Expressão Gênica , Glomerulonefrite por IGA/metabolismo , Glicerídeos , Humanos , Ligantes , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Miofibroblastos/efeitos dos fármacos , Nefrite Intersticial/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/análise , Receptor CB2 de Canabinoide/análise , Receptor CB2 de Canabinoide/genética , Rimonabanto , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima , Obstrução Ureteral/complicações , Obstrução Ureteral/metabolismo
14.
Am J Pathol ; 184(9): 2438-49, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25005449

RESUMO

Because genetic background plays a pivotal role in humans and in various experimental models, we carefully monitored its impact on glomerular pathological characteristics during experimental anti-glomerular basement membrane glomerulonephritis (anti-GBM-GN), using two leading mouse strains, 129S2/SvPas (129Sv) and C57bl/6J (B6J). These mice exhibited different severities of renal failure, hypertension, and glomerular lesions, according to their genetic background. In addition to the classic glomerular proliferative lesions, glomerular thrombotic microangiopathy (TMA) was found as a common genetic background-dependent histopathological hallmark of anti-GBM-GN, combined with hemolytic anemia and thrombocytopenia. Glomerular expression profiling, using microarrays and Western blot analysis in B6J TMA-resistant and 129Sv TMA-prone mice, demonstrated major differences in vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) 2 pathways, despite similar Vegfa expression levels. Further analysis revealed a lower basal glomerular endothelial Vegfr2 expression level in 129Sv TMA-prone mice compared with B6J TMA-resistant mice. This difference was even more pronounced during anti-GBM-GN, explaining why an exogenous VEGFA supply failed to rescue any 129Sv TMA lesions. Conversely, the systemic blocking of Vegfr2 amplified TMA lesions only in B6J mice. Herein, we specified the role that genetic background plays in determining, in particular, the level of Vegfr2 expression. We also demonstrated that glomerular Vegfr2-dependent TMA lesions are an underevaluated common hallmark of anti-GBM-GN in mice.


Assuntos
Doença Antimembrana Basal Glomerular/genética , Doença Antimembrana Basal Glomerular/patologia , Transdução de Sinais/fisiologia , Microangiopatias Trombóticas/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Doença Antimembrana Basal Glomerular/metabolismo , Western Blotting , Modelos Animais de Doenças , Imunofluorescência , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Análise Serial de Tecidos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
J Am Soc Nephrol ; 25(8): 1662-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24790179

RESUMO

Circulating levels of soluble forms of urokinase-type plasminogen activator receptor (suPAR) are generally elevated in sera from children and adults with FSGS compared with levels in healthy persons or those with other types of kidney disease. In mice lacking the gene encoding uPAR, forced increases in suPAR concentration result in FSGS-like glomerular lesions and proteinuria. However, whether overexpression of suPAR, per se, contributes to the pathogenesis of FSGS in humans remains controversial. We conducted an independent set of animal experiments in which two different and well characterized forms of recombinant suPAR produced by eukaryotic cells were administered over the short or long term to wild-type (WT) mice. In accordance with the previous study, the delivered suPARs are deposited in the glomeruli. However, such deposition of either form of suPAR in the kidney did not result in increased glomerular proteinuria or altered podocyte architecture. Our findings suggest that glomerular deposits of suPAR caused by elevated plasma levels are not sufficient to engender albuminuria.


Assuntos
Nefrite/etiologia , Podócitos/metabolismo , Podócitos/patologia , Proteinúria/etiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Camundongos Endogâmicos C57BL , Nefrite/metabolismo , Nefrite/patologia , Proteinúria/metabolismo , Proteinúria/patologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/administração & dosagem , Proteínas Recombinantes/administração & dosagem
16.
Kidney Int Rep ; 9(4): 1093-1106, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38765560

RESUMO

Introduction: During glomerular diseases, podocyte-specific pathways can modulate the intensity of histological disease and prognosis. The therapeutic targeting of these pathways could thus improve the management and prognosis of kidney diseases. The Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway, classically described in immune cells, has been recently described in detail in intrinsic kidney cells. Methods: We describe STAT5 expression in human kidney biopsies from patients with focal segmental glomerulosclerosis (FSGS) and studied mice with a podocyte-specific Stat5 deletion in experimental glomerular diseases. Results: Here, we show, for the first time, that STAT5 is activated in human podocytes in FSGS. In addition, podocyte-specific Stat5 inactivation aggravates the structural and functional alterations in a mouse model of FSGS. This could be due, at least in part, to an inhibition of autophagic flux. Finally, interleukin 15 (IL-15), a classical activator of STAT5 in immune cells, increases STAT5 phosphorylation in human podocytes, and its administration alleviates glomerular injury in vivo by maintaining autophagic flux in podocytes. Conclusion: Activating podocyte STAT5 with commercially available IL-15 represents a potential new therapeutic avenue for FSGS.

17.
FASEB J ; 26(10): 4079-91, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22751008

RESUMO

This study investigated the role of discoidin domain receptor 1 (DDR1), a collagen receptor that displays tyrosine-kinase activity, in the development of glomerulonephritis. Crescentic glomerulonephritis was induced in DDR1-deficient mice and their wild-type (WT) littermates as controls, by injection of alloimmune sheep nephrotoxic serum (NTS). Histological, functional and transcriptomic studies were performed. Glomerulonephritis produced a 17-fold increase of DDR1 expression, predominantly in glomeruli. DDR1 deletion protected NTS-treated mice against glomerular disease (proteinuria/creatininuria 5.5±1.1 vs. 13.2±0.8 g/mmol in WT, crescents 12±2 vs. 24±2% of glomeruli, urea 16±2 vs. 28±5 mM), hypertension (123±11 vs. 157±8 mmHg), and premature death (70 vs. 10% survival) (all P<0.05). Reciprocal stimulation between DDR1 and interleukin-1b expression in vivo and in cultured podocytes suggested a positive feed-back loop between DDR1 and inflammation. In NTS-treated WT mice, administration of DDR1-specific antisense oligodeoxynucleotides decreased DDR1 expression (-56%) and protected renal function and structure, including nephrin expression (4.2±1.4 vs. 0.9±0.4 arbitrary units, P<0.05), compared to control mice receiving scrambled oligodeoxynucleotides. The therapeutic potential of this approach was reinforced by the observation of increased DDR1 expression in glomeruli of patients with lupus nephritis and Goodpasture's syndrome. These results prompt further interest in DDR1 blockade strategies, especially in the treatment of glomerulonephritis.


Assuntos
Glomerulonefrite/prevenção & controle , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Mitogênicos/metabolismo , Animais , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Nitrogênio da Ureia Sanguínea , Western Blotting , Receptores com Domínio Discoidina , Feminino , Imunofluorescência , Glomerulonefrite/genética , Glomerulonefrite/urina , Humanos , Imuno-Histoquímica , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteinúria/genética , Proteinúria/urina , Receptores Proteína Tirosina Quinases/genética , Receptores Mitogênicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Kidney Int ; 82(4): 428-35, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22513823

RESUMO

The renal urothelium, the monolayered epithelium that covers the papilla, is the direct target of increased pressure during obstruction, yet most studies have mainly focused on tubules, fibroblasts, and inflammatory cells. We studied this epithelium in a unilateral ureteral obstruction mouse mode land found that it was disrupted and had broken tight junctions, enlarged intercellular space, with loss of apicaluroplakins, and marginal lumen desquamation. Shortly after obstruction these urothelial cells proliferated, peaking at day 2. By day 14, the renal urothelium was transformed into a multilayered barrier with newly synthesized uroplakins including the de novo induction of uroplakin II. This proliferation was found to be fibroblast growth factor (FGF)dependent. Renal urothelial cells constitutively express the FGF receptor 2, and obstruction activated the receptor by phosphorylation. Treatment with FGF receptor 2-antisense or vitamin A (an inhibitor of the MAP kinase in the FGFR2 pathway) decreased renal urothelial cell proliferation. Among known FGF receptor 2 ligands, only FGF7 was upregulated.Infusion of FGF7 into control mice caused the formation of a multilayered structure at 7 days, resembling the urothelium 14 days following obstruction. Thus, the pressure/stretching of renal monolayered urothelial cells is a very efficient trigger for proliferation, causing the formation of a bladder-like multistratified barrier with enhanced apical uroplakin plaques. Presumably, this ensures efficient barrier protection and repair.


Assuntos
Diferenciação Celular , Proliferação de Células , Transdiferenciação Celular , Rim/patologia , Obstrução Ureteral/patologia , Bexiga Urinária/patologia , Urotélio/patologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transdiferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Fator 7 de Crescimento de Fibroblastos/administração & dosagem , Fator 7 de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Mecanotransdução Celular , Camundongos , Camundongos Endogâmicos C57BL , Oligonucleotídeos Antissenso/metabolismo , Fenótipo , Fosforilação , Pressão , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Estresse Mecânico , Fatores de Tempo , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/metabolismo , Uroplaquinas/metabolismo , Urotélio/efeitos dos fármacos , Urotélio/metabolismo , Vitamina A/farmacologia
19.
Kidney Int ; 81(12): 1226-38, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22418977

RESUMO

Thrombospondin-1 (TSP-1) is an endogenous activator of transforming growth factor-ß (TGF-ß), and an anti-angiogenic factor, which may prevent kidney repair. Here we investigated whether TSP-1 is involved in the development of chronic kidney disease using rats with unilateral ureteral obstruction, a well-known model to study renal fibrosis. Obstruction of 10 days duration induced inflammation, tubular cell atrophy, dilation, apoptosis, and proliferation, leading to interstitial fibrosis. TSP-1 expression was increased in parallel to that of collagen III and TGF-ß. Relief of the obstruction at day 10 produced a gradual improvement in renal structure and function, the reappearance of peritubular capillaries, and restoration of renal VEGF content over a 7- to 15-day post-relief period. TSP-1 expression decreased in parallel with that of TGF-ß1 and collagen III. Mice in which the TSP-1 gene was knocked out displayed less inflammation and had better preservation of renal tissue and the peritubular capillary network compared to wild-type mice. Additional studies showed that the inflammatory effect of TSP-1 was mediated, at least in part, by monocyte chemoattractant protein-1 and activation of the Th17 pathway. Thus, TSP-1 is an important profibrotic and inflammatory mediator of renal disease. Blockade of its action may be a treatment against the development of chronic kidney disease.


Assuntos
Mediadores da Inflamação/metabolismo , Nefropatias/etiologia , Rim/metabolismo , Nefrite/etiologia , Trombospondina 1/metabolismo , Obstrução Ureteral/complicações , Animais , Apoptose , Atrofia , Capilares/metabolismo , Capilares/patologia , Proliferação de Células , Quimiocina CCL2/metabolismo , Doença Crônica , Colágeno Tipo III/metabolismo , Modelos Animais de Doenças , Fibrose , Regulação da Expressão Gênica , Rim/irrigação sanguínea , Rim/patologia , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos , Camundongos Knockout , Nefrectomia , Nefrite/genética , Nefrite/metabolismo , Nefrite/patologia , Nefrite/prevenção & controle , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Trombospondina 1/deficiência , Trombospondina 1/genética , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Am J Pathol ; 179(1): 83-91, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21640971

RESUMO

The interactions between tubulointerstitial infiltrating cells and the extracellular matrix play an important role in regulating renal fibrosis. Discoidin domain receptor 1 (DDR1) is a nonintegrin tyrosine kinase receptor for collagen implicated in cell adhesion, proliferation, and extracellular matrix remodeling. We have previously demonstrated that transgenic mice lacking DDR1 are protected from hypertension-associated renal fibrosis. The purpose of this study was to determine the role of DDR1 in renal inflammation and fibrosis related to primitive tubulointerstitial injury. After 12 days of unilateral ureteral obstruction (UUO), kidney histopathologic and real-time quantitative PCR analyses were performed in DDR1(-/-) and wild-type mice. DDR1 expression was strongly increased in the obstructed kidney. Wild-type mice developed important perivascular and interstitial inflammation and fibrosis. In comparison, DDR1(-/-) mice displayed reduced accumulation of fibrillar collagen and transforming growth factor ß expression. F4/80(+) cell count and proinflammatory cytokines were remarkably blunted in DDR1(-/-) obstructed kidneys. Leukocyte rolling and adhesion evaluated by intravital microscopy were not different between DDR1(-/-) and wild-type mice. Importantly, macrophages isolated from DDR1(-/-) mice presented similar M1/M2 polarization but displayed impaired migration in response to monocyte chemoattractant protein-1. Together, these data suggest that DDR1 plays an important role in the pathogenesis of renal disease via enhanced inflammation. Inhibition of DDR1 expression or activity may represent a novel therapeutic target against the progression of renal diseases.


Assuntos
Movimento Celular , Fibrose/etiologia , Inflamação/etiologia , Nefropatias/complicações , Receptores Proteína Tirosina Quinases/fisiologia , Animais , Western Blotting , Adesão Celular , Proliferação de Células , Células Cultivadas , Receptor com Domínio Discoidina 1 , Fibrose/patologia , Humanos , Inflamação/patologia , Nefropatias/patologia , Leucócitos/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa