Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Kidney Int ; 98(3): 601-614, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32739209

RESUMO

Transforming growth factor-ß (TGF-ß) is a central mediator of diabetic nephropathy. The effect of TGF-ß, mediated by the type I TGF-ß receptor, ALK5, and subsequent Smad2/3 activation results in podocyte apoptosis and loss. Previously, we demonstrated that the genetic deletion of the BMP and Activin Membrane-Bound Inhibitor (BAMBI), a negative modulator TGF-ß signaling, accelerates diabetic nephropathy in mice. This was associated with heightened ALK1-mediated activation of Smad1/5 in the glomerular endothelial cells (ECs). Therefore, to evaluate the glomerular cell-specific effects of TGF-ß in diabetic nephropathy we examined the effects of the podocyte- or EC-specific loss of Bambi (Pod-Bambi-/- or EC-Bambi-/-) in streptozotocin-induced diabetic mice with endothelial nitric oxide synthase deficiency. Interestingly, although hyperglycemia and body weight loss were similar in all groups of diabetic mice, significant hypertension was present only in the diabetic EC-Bambi-/- mice. While the podocyte or EC-specific loss of BAMBI both accelerated the progression of diabetic nephropathy, the worsened podocyte injury and loss observed in the diabetic Pod-Bambi-/- mice were associated with enhanced Smad3 activation. Increased Smad1/5 activation and EC proliferation were apparent only in the glomeruli of diabetic EC-Bambi-/- mice. The enhanced Smad1/5 activation in diabetic EC-Bambi-/- mice was associated with increased glomerular expression of plasmalemma vesicle-associated protein, pointing to the involvement of immature or dedifferentiated glomerular ECs in diabetic nephropathy. Notably, diabetic EC-Bambi-/- mice displayed podocyte injury and loss that were comparable to diabetic Pod-Bambi-/- mice. Thus, our results highlight the glomerular cell-specific contribution of TGF-ß signaling and the intricate cross-talk between injured glomerular cells in the progression of diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Podócitos , Animais , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/genética , Células Endoteliais , Camundongos , Fator de Crescimento Transformador beta , Fatores de Crescimento Transformadores
2.
Nephrol Dial Transplant ; 35(4): 564-572, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30879039

RESUMO

Crescentic glomerulonephritis (cGN) comprises three main types according to the pathogenesis and immunofluorescence patterns: anti-glomerular basement membrane antibody cGN, vasculitis-associated cGN and post-infectious immune complex cGN. In this brief review of the immune-pathogenesis of cGN, the focus is mainly on the role of CD8+ T cells in the progression of cGN. Under control conditions, Bowman's capsule (BC) provides a protected immunological niche by preventing access of cytotoxic CD8+ T cells to Bowman's space and thereby podocytes. Even in experimental nephrotoxic nephritis, leukocytes accumulate around the glomeruli, but remain outside of BC, as long as the latter remains intact. However, when and where breaches in BC occur, the inflammatory cells can gain access to and destroy podocytes, thus converting cGN into rapidly progressive glomerulonephritis (RPGN). These conclusions also apply to human cGN, where biopsies show that loss of BC integrity is associated with RPGN and progression to end-stage kidney disease. We propose a two-hit hypothesis for the role of cytotoxic CD8+ T cells in the progression of cGN. The initial insult occurs in response to the immune complex formation or deposition, resulting in local capillary and podocyte injury (first hit). The injured podocytes release neo-epitopes, eventually causing T-cell activation and migration to the glomerulus. Upon generation of breaches in BC, macrophages and CD8+ T cells can now gain access to the glomerular space and destroy neo-epitope expressing podocytes (second hit), resulting in RPGN. While further investigation will be required to test this hypothesis, future therapeutic trials should consider targeting of CD8+ T cells in the therapy of progressive cGN.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Glomerulonefrite/etiologia , Linfócitos T CD8-Positivos/patologia , Progressão da Doença , Glomerulonefrite/patologia , Humanos
3.
J Am Soc Nephrol ; 30(4): 533-545, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30846559

RESUMO

BACKGROUND: Recent single-cell RNA sequencing (scRNA-seq) analyses have offered much insight into cell-specific gene expression profiles in normal kidneys. However, in diseased kidneys, understanding of changes in specific cells, particularly glomerular cells, remains limited. METHODS: To elucidate the glomerular cell-specific gene expression changes in diabetic kidney disease, we performed scRNA-seq analysis of isolated glomerular cells from streptozotocin-induced diabetic endothelial nitric oxide synthase (eNOS)-deficient (eNOS-/-) mice and control eNOS-/- mice. RESULTS: We identified five distinct cell populations, including glomerular endothelial cells, mesangial cells, podocytes, immune cells, and tubular cells. Using scRNA-seq analysis, we confirmed the expression of glomerular cell-specific markers and also identified several new potential markers of glomerular cells. The number of immune cells was significantly higher in diabetic glomeruli compared with control glomeruli, and further cluster analysis showed that these immune cells were predominantly macrophages. Analysis of differential gene expression in endothelial and mesangial cells of diabetic and control mice showed dynamic changes in the pattern of expressed genes, many of which are known to be involved in diabetic kidney disease. Moreover, gene expression analysis showed variable responses of individual cells to diabetic injury. CONCLUSIONS: Our findings demonstrate the ability of scRNA-seq analysis in isolated glomerular cells from diabetic and control mice to reveal dynamic changes in gene expression in diabetic kidneys, with variable responses of individual cells. Such changes, which might not be apparent in bulk transcriptomic analysis of glomerular cells, may help identify important pathophysiologic factors contributing to the progression of diabetic kidney disease.


Assuntos
Nefropatias Diabéticas/genética , Glomérulos Renais/metabolismo , Túbulos Renais/metabolismo , Macrófagos/metabolismo , RNA Citoplasmático Pequeno/análise , Transcriptoma , Animais , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/etiologia , Células Endoteliais/metabolismo , Glomérulos Renais/patologia , Túbulos Renais/patologia , Células Mesangiais/metabolismo , Camundongos , Podócitos , Análise de Sequência de RNA , Análise de Célula Única
4.
J Am Soc Nephrol ; 30(4): 546-562, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30858225

RESUMO

BACKGROUND: Glomerular endothelial dysfunction and neoangiogenesis have long been implicated in the pathogenesis of diabetic kidney disease (DKD). However, the specific molecular pathways contributing to these processes in the early stages of DKD are not well understood. Our recent transcriptomic profiling of glomerular endothelial cells identified a number of proangiogenic genes that were upregulated in diabetic mice, including leucine-rich α-2-glycoprotein 1 (LRG1). LRG1 was previously shown to promote neovascularization in mouse models of ocular disease by potentiating endothelial TGF-ß/activin receptor-like kinase 1 (ALK1) signaling. However, LRG1's role in the kidney, particularly in the setting of DKD, has been unclear. METHODS: We analyzed expression of LRG1 mRNA in glomeruli of diabetic kidneys and assessed its localization by RNA in situ hybridization. We examined the effects of genetic ablation of Lrg1 on DKD progression in unilaterally nephrectomized, streptozotocin-induced diabetic mice at 12 and 20 weeks after diabetes induction. We also assessed whether plasma LRG1 was associated with renal outcome in patients with type 2 diabetes. RESULTS: LRG1 localized predominantly to glomerular endothelial cells, and its expression was elevated in the diabetic kidneys. LRG1 ablation markedly attenuated diabetes-induced glomerular angiogenesis, podocyte loss, and the development of diabetic glomerulopathy. These improvements were associated with reduced ALK1-Smad1/5/8 activation in glomeruli of diabetic mice. Moreover, increased plasma LRG1 was associated with worse renal outcome in patients with type 2 diabetes. CONCLUSIONS: These findings identify LRG1 as a potential novel pathogenic mediator of diabetic glomerular neoangiogenesis and a risk factor in DKD progression.


Assuntos
Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Glicoproteínas/sangue , Glicoproteínas/genética , Glomérulos Renais/metabolismo , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animais , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/fisiopatologia , Progressão da Doença , Células Endoteliais/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Taxa de Filtração Glomerular , Glicoproteínas/metabolismo , Humanos , Falência Renal Crônica/etiologia , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/patologia , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica/genética , Podócitos , Transdução de Sinais/genética , Proteínas Smad/metabolismo
5.
Kidney Int ; 94(2): 326-345, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29861058

RESUMO

Endothelial dysfunction promotes the pathogenesis of diabetic nephropathy (DN), which is considered to be an early event in disease progression. However, the molecular changes associated with glomerular endothelial cell (GEC) injury in early DN are not well defined. Most gene expression studies have relied on the indirect assessment of GEC injury from isolated glomeruli or renal cortices. Here, we present transcriptomic analysis of isolated GECs, using streptozotocin-induced diabetic wildtype (STZ-WT) and diabetic eNOS-null (STZ-eNOS-/-) mice as models of mild and advanced DN, respectively. GECs of both models in comparison to their respective nondiabetic controls showed significant alterations in the regulation of apoptosis, oxidative stress, and proliferation. The extent of these changes was greater in STZ-eNOS-/- than in STZ-WT GECs. Additionally, genes in STZ-eNOS-/- GECs indicated further dysregulation in angiogenesis and epigenetic regulation. Moreover, a biphasic change in the number of GECs, characterized by an initial increase and subsequent decrease over time, was observed only in STZ-eNOS-/- mice. This is consistent with an early compensatory angiogenic process followed by increased apoptosis, leading to an overall decrease in GEC survival in DN progression. From the genes altered in angiogenesis in STZ-eNOS-/- GECs, we identified potential candidate genes, Lrg1 and Gpr56, whose function may augment diabetes-induced angiogenesis. Thus, our results support a role for GEC in DN by providing direct evidence for alterations of GEC gene expression and molecular pathways. Candidate genes of specific pathways, such as Lrg1 and Gpr56, can be further explored for potential therapeutic targeting to mitigate the initiation and progression of DN.


Assuntos
Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/patologia , Células Endoteliais/metabolismo , Glomérulos Renais/patologia , Neovascularização Patológica/patologia , Animais , Linhagem Celular , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/genética , Células Endoteliais/patologia , Epigênese Genética , Perfilação da Expressão Gênica , Humanos , Glomérulos Renais/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/genética , Óxido Nítrico Sintase Tipo III/genética , Estresse Oxidativo , Transdução de Sinais/genética , Estreptozocina/toxicidade , Regulação para Cima
6.
Kidney Int ; 92(3): 533-536, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28807257

RESUMO

The glomerular filtration barrier (GFB) is characterized by a very high hydraulic permeability, combined with a marked permselectivity that excludes macromolecules such as albumin. Thus, the GFB retains most of the plasma proteins, with only 0.06% of albumin getting across the basement membrane. The GFB consists of 3 layers: fenestrated endothelial cells, the glomerular basement membrane, and podocytes. Injury to any of these components can result in the development of proteinuria. The contribution of the major components of the GFB has recently been reexamined and is discussed in the context of our past and present understanding.


Assuntos
Membrana Basal Glomerular , Podócitos , Células Endoteliais , Barreira de Filtração Glomerular , Humanos , Glomérulos Renais , Proteinúria
7.
Kidney Int ; 89(4): 734-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26994568

RESUMO

The goal of "precision medicine" is to characterize diseases based on the underlying molecular biology, in order to identify specific biomarkers and therapeutic targets that will ultimately improve clinical outcomes. The nephrology research community has developed a strong foundation for precision medicine, and recent publications demonstrate the feasibility of this approach to identify potential biomarkers and therapeutic targets in chronic kidney disease.


Assuntos
Biomarcadores/urina , Nefrologia/tendências , Medicina de Precisão , Insuficiência Renal Crônica/urina , Animais , Humanos , Insuficiência Renal Crônica/terapia
8.
Am J Physiol Renal Physiol ; 306(11): F1372-80, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24598802

RESUMO

Mitochondrial dysfunction is increasingly recognized as contributing to glomerular diseases, including those secondary to mitochondrial DNA (mtDNA) mutations and deletions. Mitochondria maintain cellular redox and energy homeostasis and are a major source of intracellular reactive oxygen species (ROS) production. Mitochondrial ROS accumulation may contribute to stress-induced mitochondrial dysfunction and apoptosis and thereby to glomerulosclerosis. In mice, deletion of the gene encoding Mpv17 is associated with glomerulosclerosis, but the underlying mechanism remains poorly defined. Here we report that Mpv17 localizes to mitochondria of podocytes and its expression is reduced in several glomerular injury models and in human focal segmental glomerulosclerosis (FSGS) but not in minimal change disease. Using models of mild or severe nephrotoxic serum nephritis (NTSN) in Mpv17(+/+) wild-type (WT) and Mpv17(-/-) knockout mice, we found that Mpv17 deficiency resulted in increased proteinuria (mild NTSN) and renal insufficiency (severe NTSN) compared with WT. These lesions were associated with increased mitochondrial ROS generation and mitochondrial injury such as oxidative DNA damage. In vitro, podocytes with loss of Mpv17 function were characterized by increased susceptibility to apoptosis and ROS injury including decreased mitochondrial function, loss of mtDNA content, and change in mitochondrial configuration. In summary, the inner mitochondrial membrane protein Mpv17 in podocytes is essential for the maintenance of mitochondrial homeostasis and protects podocytes against oxidative stress-induced injury both in vitro and in vivo.


Assuntos
Apoptose/fisiologia , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Nefrite/metabolismo , Estresse Oxidativo/fisiologia , Podócitos/metabolismo , Animais , Modelos Animais de Doenças , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Camundongos , Camundongos Transgênicos , Mitocôndrias/patologia , Nefrite/patologia , Podócitos/patologia , Proteinúria/metabolismo , Proteinúria/patologia , Espécies Reativas de Oxigênio/metabolismo
9.
Kidney Int ; 85(3): 499-501, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24583981

RESUMO

A soluble proteinuric substance has been postulated as a cause of primary focal segmental glomerulosclerosis (FSGS), and one candidate soluble urokinase receptor (suPAR). Evaluation of published results shows that serum suPAR determinations by current methodology do not reliably distinguish FSGS from other proteinuric glomerular diseases and serum suPAR cannot currently be considered a valid biomarker for primary or secondary FSGS. However, this should not discourage further research on potential roles of suPAR in proteinuric renal disease, including FSGS.


Assuntos
Glomerulonefrite/sangue , Glomerulosclerose Segmentar e Focal/sangue , Glomerulosclerose Segmentar e Focal/diagnóstico , Síndrome Nefrótica/sangue , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Feminino , Humanos , Masculino
10.
J Immunol ; 188(10): 5106-15, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22490864

RESUMO

Renal fibrosis and inflammation are associated with hypoxia, and tissue pO(2) plays a central role in modulating the progression of chronic kidney disease. Key mediators of cellular adaptation to hypoxia are hypoxia-inducible factor (HIF)-1 and -2. In the kidney, they are expressed in a cell type-specific manner; to what degree activation of each homolog modulates renal fibrogenesis and inflammation has not been established. To address this issue, we used Cre-loxP recombination to activate or to delete both Hif-1 and Hif-2 either globally or cell type specifically in myeloid cells. Global activation of Hif suppressed inflammation and fibrogenesis in mice subjected to unilateral ureteral obstruction, whereas activation of Hif in myeloid cells suppressed inflammation only. Suppression of inflammatory cell infiltration was associated with downregulation of CC chemokine receptors in renal macrophages. Conversely, global deletion or myeloid-specific inactivation of Hif promoted inflammation. Furthermore, prolonged hypoxia suppressed the expression of multiple inflammatory molecules in noninjured kidneys. Collectively, we provide experimental evidence that hypoxia and/or myeloid cell-specific HIF activation attenuates renal inflammation associated with chronic kidney injury.


Assuntos
Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Células Mieloides/imunologia , Células Mieloides/patologia , Obstrução Ureteral/imunologia , Obstrução Ureteral/patologia , Injúria Renal Aguda/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Modelos Animais de Doenças , Fibrose/imunologia , Fibrose/prevenção & controle , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células Mieloides/metabolismo , Cultura Primária de Células , Obstrução Ureteral/genética
11.
J Am Soc Nephrol ; 24(5): 787-99, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23539761

RESUMO

TGFß signaling plays a central role in the development of acute and chronic kidney diseases. Previous in vivo studies involved systemic alteration of TGFß signaling, however, limiting conclusions about the direct role of TGFß in tubular cell injury. Here, we generated a double transgenic mouse that inducibly expresses a ligand-independent constitutively active TGFß receptor type 1 (TßR1) kinase specifically in tubular epithelial cells, with expression restricted by the Pax8 promoter. In this model, activation of TGFß signaling in the tubular epithelium alone was sufficient to cause AKI characterized by marked tubular cell apoptosis and necrosis, oxidative stress, dedifferentiation and regenerative cell proliferation, reduced renal function, and interstitial accumulation of inflammatory cells. This tubular injury was associated with mitochondrial-derived generation of reactive oxygen species (ROS), but cell damage and apoptosis were partially independent of mitochondrial-derived ROS. TßR1 signaling-induced tubular injury also associated with significant leukocyte infiltration consisting of F4/80(+) macrophages, CD11c(+) F4/80(+) dendritic cells, CD11c(+) F4/80(-) Ly6C(high) dendritic cells/monocytes, and T cells. Inhibition of mitochondrial-derived ROS significantly reduced accumulation of CD11c(+) F4/80(+) dendritic cells and T cells, suggesting a role for ROS in the activation and recruitment of the adaptive immune response to tubular injury. Taken together, these results suggest that TGFß signaling in the tubular epithelium alone is sufficient to cause acute tubular injury and inflammation; therefore, TGFß may be a mechanistic link between acute injury and chronic progression of kidney disease.


Assuntos
Túbulos Renais/patologia , Nefrite Intersticial/etiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Doença Aguda , Imunidade Adaptativa , Animais , Apoptose , Antígeno CD11c/análise , Proliferação de Células , Quimiocina CCL2/fisiologia , Células Dendríticas/fisiologia , Células Epiteliais/metabolismo , Leucócitos/fisiologia , Macrófagos/fisiologia , Camundongos , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Linfócitos T/fisiologia
13.
Kidney Int ; 81(1): 100-11, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21918500

RESUMO

Polyomavirus-associated nephropathy (PVAN) is a significant complication after kidney transplantation, often leading to premature graft loss. In order to identify antiviral responses of the renal tubular epithelium, we studied activation of the viral DNA and the double-stranded RNA (dsRNA) sensors Toll-like receptor 3 (TLR3) and retinoic acid inducible gene-I (RIG-I) in allograft biopsy samples of patients with PVAN, and in human collecting duct cells in culture after stimulation by the dsRNA mimic polyriboinosinic:polyribocytidylic acid (poly(I:C)), cytokines, or infection with BK virus. Double staining using immunofluorescence for BK virus and TLR3 showed strong signals in epithelial cells of distal cortical tubules and the collecting duct. In biopsies microdissected to isolate tubulointerstitial lesions, TLR3 but not RIG-I mRNA expression was found to be increased in PVAN. Collecting duct cells in culture expressed TLR3 intracellularly, and activation of TLR3 and RIG-I by poly(I:C) enhanced expression of cytokine, chemokine, and IFN-ß mRNA. This inflammatory response could be specifically blocked by siRNA to TLR3. Finally, infection of the collecting duct cells with BK virus enhanced the expression of cytokines and chemokines. This led to an efficient antiviral immune response with TLR3 and RIG-I upregulation without activation of IL-1ß or components of the inflammasome pathway. Thus, PVAN activation of innate immune defense mechanisms through TLR3 is involved in the antiviral and anti-inflammatory response leading to the expression of proinflammatory cytokines and chemokines.


Assuntos
Vírus BK , Imunidade Inata , Nefropatias/etiologia , Nefropatias/imunologia , Transplante de Rim/efeitos adversos , Infecções por Polyomavirus/imunologia , Infecções Tumorais por Vírus/imunologia , Vírus BK/imunologia , Vírus BK/patogenicidade , Sequência de Bases , Células Cultivadas , Quimiocinas/genética , Citocinas/genética , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Técnicas de Silenciamento de Genes , Humanos , Nefropatias/genética , Infecções por Polyomavirus/complicações , Infecções por Polyomavirus/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Receptores Imunológicos , Receptor 3 Toll-Like/antagonistas & inibidores , Receptor 3 Toll-Like/genética , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/genética
15.
Eur J Immunol ; 40(1): 267-78, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19830734

RESUMO

The chemokine (C-C motif) receptor 5 (CCR5) has been implicated in experimental and clinical allograft rejection. To dissect the function of CCR5 in acute and chronic renal allograft rejection, bilaterally nephrectomized WT and Ccr5-/- C57BL/6 mice were used as recipients of WT BALB/c renal allografts and analyzed 7 and 42 days after transplantation. Lesion scores (glomerular damage, vascular rejection, tubulointerstitial inflammation) and numbers of CD4+, CD8+, CD11c+ and alpha smooth muscle actin (alphaSMA)+ cells were reduced in allografts from Ccr5-/- recipients during the chronic phase. Increasing creatinine levels indicated deterioration of allograft function over time. While mRNA expression of Th1-associated markers decreased between 7 and 42 days, Th2-associated markers increased. Markers for alternatively activated macrophages (arginase 1, chitinase 3-like 3, resistin-like alpha, mannose receptor, C type 1), were strongly upregulated (mRNA and/or protein level) only in allografts from Ccr5-/- recipients at 42 days. Ccr5 deficiency shifted intragraft immune responses during the chronic phase towards the Th2 type and led to accumulation of alternatively activated macrophages. Additionally, splenocytes from unchallenged Ccr5-/- mice showed significantly increased arginase 1 and mannose receptor 1 mRNA levels, suggesting constitutive alternative activation of splenic macrophages. We conclude that Ccr5 deficiency favors alternative macrophage activation. This finding may be relevant for other inflammatory diseases that involve macrophage activation and may also influence future therapeutic strategies targeting CCR5.


Assuntos
Sobrevivência de Enxerto , Transplante de Rim/imunologia , Macrófagos/imunologia , Receptores CCR5/imunologia , Animais , Polaridade Celular , Regulação da Expressão Gênica , Rim/fisiologia , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CCR5/deficiência , Fatores de Tempo , Transplante Homólogo/imunologia , Resultado do Tratamento
16.
Am J Pathol ; 176(2): 594-607, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20019191

RESUMO

In the kidney, hypoxia contributes to tubulointerstitial fibrosis, but little is known about its implications for glomerular damage and glomerulosclerosis. Chronic hypoxia was hypothesized to be involved in nephrosclerosis (NSC) or "hypertensive nephropathy." In the present study genome-wide expression data from microdissected glomeruli were studied to examine the role of hypoxia in glomerulosclerosis of human NSC. Functional annotation analysis revealed prominent regulation of hypoxia-associated biological processes in NSC, including angiogenesis, fibrosis, and inflammation. Glomerular expression levels of a majority of genes regulated by the hypoxia-inducible factors (HIFs) were significantly altered in NSC. Among these HIF targets, chemokine C-X-C motif receptor 4 (CXCR4) was prominently induced. Glomerular CXCR4 mRNA induction was confirmed by quantitative RT-PCR in an independent cohort with NSC but not in those with other glomerulopathies. By immunohistological analysis, CXCR4 showed enhanced positivity in podocytes in NSC biopsy specimens. This CXCR4 positivity was associated with nuclear localization of HIF1alpha only in podocytes of NSC, indicating transcriptional activity of HIF. As the CXCR4 ligand CXCL12/SDF-1 is constitutively expressed in podocytes, autocrine signaling may contribute to NSC. In addition, a blocking CXCR4 antibody caused significant inhibition of wound closure by podocytes in an in vitro scratch assay. These data support a role for CXCR4/CXCL12 in human NSC and indicate that hypoxia not only is involved in tubulointerstitial fibrosis but also contributes to glomerular damage in NSC.


Assuntos
Hipóxia/complicações , Nefropatias/etiologia , Nefroesclerose/complicações , Adolescente , Adulto , Idoso , Animais , Estudos de Casos e Controles , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Humanos , Hipóxia/genética , Nefropatias/genética , Nefropatias/patologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Nefroesclerose/genética , Análise de Sequência com Séries de Oligonucleotídeos , Oxigênio/farmacologia , Adulto Jovem
17.
Nat Med ; 9(1): 40-6, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12461524

RESUMO

Selective skewing of autoreactive interferon-gamma (IFN-gamma)-producing T helper cells (Th1) toward an interleukin-4 (IL-4)-producing (Th2) phenotype can in experimental animals alleviate autoimmune disease without inducing general immunosuppression. In a prospective dose escalation study, we assessed treatment with human IL-4 (rhuIL-4) in 20 patients with severe psoriasis. The therapy was well tolerated, and within six weeks all patients showed decreased clinical scores and 15 improved more than 68%. Stable reduction of clinical scores was significantly better at 0.2-0.5 microg rhuIL-4 than at < or =0.1 microg rhuIL-4 (P = 0.009). In psoriatic lesions, treatment with 0.2-0.5 microg/kg rhuIL-4 reduced the concentrations of IL-8 and IL-19, two cytokines directly involved in psoriasis; the number of chemokine receptor CCR5+ Th1 cells; and the IFN-gamma/IL-4 ratio. In the circulation, 0.2-0.5 microg/kg rhuIL-4 increased the number of IL-4+CD4+ T cells two- to three-fold. Thus, IL-4 therapy can induce Th2 differentiation in human CD4+ T cells and has promise as a potential treatment for psoriasis, a prototypic Th1-associated autoimmune disease.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Interleucina-4/imunologia , Interleucina-4/uso terapêutico , Psoríase/tratamento farmacológico , Psoríase/imunologia , Células Th2/imunologia , Adolescente , Adulto , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Pré-Escolar , Feminino , Humanos , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-18/metabolismo , Interleucina-4/administração & dosagem , Interleucina-4/farmacologia , Interleucinas , Masculino , Pessoa de Meia-Idade , Fenótipo , Psoríase/patologia , Psoríase/fisiopatologia , Receptores CCR5/metabolismo , Índice de Gravidade de Doença , Pele/citologia , Pele/efeitos dos fármacos , Pele/patologia , Células Th1/citologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/metabolismo
18.
Kidney Int ; 78(1): 29-37, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20428100

RESUMO

Inflammation is the immune system's response to infectious or noninfectious sources of danger. Danger recognition is facilitated by various innate immune receptor families including the Toll-like receptors (TLRs), which detect danger signals in extracellular and intracellular compartments. It is an evolving concept that renal damage triggers intrarenal inflammation by immune recognition of molecules that are being released by dying cells. Such danger-associated molecules act as immunostimulatory agonists to TLRs and other innate immune receptors and induce cytokine and chemokine secretion, leukocyte recruitment, and tissue remodeling. As a new entry to this concept, autophagy allows stressed cells to reduce intracellular microorganisms, protein aggregates, and cellular organelles by moving and subsequently digesting them in autophagolysosomes. Within the autophagolysosome, endogenous molecules and danger-associated molecules may be presented to TLRs or loaded onto the major histocompatibility complex and presented as autoantigens. Here we discuss the current evidence for the danger signaling concept in autoimmune kidney injury and propose that autophagy-related processing of self-proteins provides a source of immunostimulatory molecules and autoantigens. A better understanding of danger signaling should enable us to unravel yet unknown triggers for renal immunopathology and progressive kidney disease.


Assuntos
Rim/imunologia , Receptores Toll-Like/imunologia , Animais , Autofagia/imunologia , Humanos , Infecções/imunologia , Infecções/metabolismo , Infecções/patologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Rim/metabolismo , Rim/patologia , Nefropatias/imunologia , Nefropatias/metabolismo , Nefropatias/patologia , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/metabolismo
19.
Am J Pathol ; 175(1): 119-31, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19498001

RESUMO

The Duffy antigen/receptor for chemokines (DARC) is a chemokine-binding protein that is expressed on erythrocytes and renal endothelial cells. DARC-mediated endothelial transcytosis of chemokines may facilitate the renal recruitment of macrophages and T cells, as has been suggested for neutrophils. We studied the role of Darc in two mouse models of prolonged renal inflammation, one that primarily involves the tubulointerstitium (unilateral ureteral obstruction), and one that requires an adaptive immune response that leads to glomerulonephritis (accelerated nephrotoxic nephritis). Renal expression of Darc and its ligands was increased in both models. Leukocytes effectively infiltrated obstructed kidneys in Darc-deficient mice with pronounced T-cell infiltration at early time points. Development of interstitial fibrosis was comparable in both genotypes. Nephrotoxic nephritis was inducible in Darc-deficient mice, with both an increased humoral immune response and functional impairment during the early phase of disease. Leukocytes efficiently infiltrated kidneys of Darc-deficient mice, with increased cell numbers at early but not late time points. Taken together, renal inflammation developed more rapidly in DARC-deficient mice, without affecting the extent of renal injury at later time points. Thus, genetic elimination of Darc in mice does not prevent the development of renal infiltrates and may even enhance such development during the early phases of interstitial and glomerular diseases in mouse models of prolonged renal inflammation.


Assuntos
Quimiotaxia de Leucócito , Sistema do Grupo Sanguíneo Duffy , Glomerulonefrite , Macrófagos , Receptores de Superfície Celular , Linfócitos T , Animais , Camundongos , Quimiocinas/imunologia , Quimiotaxia de Leucócito/imunologia , Sistema do Grupo Sanguíneo Duffy/genética , Sistema do Grupo Sanguíneo Duffy/imunologia , Ensaio de Imunoadsorção Enzimática , Fibrose , Citometria de Fluxo , Glomerulonefrite/imunologia , Glomerulonefrite/patologia , Processamento de Imagem Assistida por Computador , Imunoglobulina G/sangue , Imuno-Histoquímica , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/imunologia
20.
Am J Pathol ; 174(6): 2073-85, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19465643

RESUMO

Although chronic kidney disease (CKD) is common, only a fraction of CKD patients progress to end-stage renal disease. Molecular predictors to stratify CKD populations according to their risk of progression remain undiscovered. Here we applied transcriptional profiling of kidneys from transforming growth factor-beta1 transgenic (Tg) mice, characterized by heterogeneity of kidney disease progression, to identify 43 genes that discriminate kidneys by severity of glomerular apoptosis before the onset of tubulointerstitial fibrosis in 2-week-old animals. Among the genes examined, 19 showed significant correlation between mRNA expression in uninephrectomized left kidneys at 2 weeks of age and renal disease severity in right kidneys of Tg mice at 4 weeks of age. Gene expression profiles of human orthologs of the 43 genes in kidney biopsies were highly significantly related (R(2) = 0.53; P < 0.001) to the estimated glomerular filtration rates in patients with CKD stages I to V, and discriminated groups of CKD stages I/II and III/IV/V with positive and negative predictive values of 0.8 and 0.83, respectively. Protein expression patterns for selected genes were successfully validated by immunohistochemistry in kidneys of Tg mice and kidney biopsies of patients with IgA nephropathy and CKD stages I to V, respectively. In conclusion, we developed novel mRNA and protein expression signatures that predict progressive renal fibrosis in mice and may be useful molecular predictors of CKD progression in humans.


Assuntos
Perfilação da Expressão Gênica , Nefropatias/genética , Nefropatias/patologia , Animais , Análise por Conglomerados , Progressão da Doença , Expressão Gênica , Humanos , Imuno-Histoquímica , Nefropatias/metabolismo , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Risco , Transcrição Gênica , Fator de Crescimento Transformador beta1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA