Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Kidney Int ; 105(2): 312-327, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37977366

RESUMO

Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Identifying the molecular and genetic regulators unique to nephron segments that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. Here, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI and found them upregulated after severe AKI and correlated with chronic injury. Surprisingly, proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to pre-conditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of proximal tubule cells in the S3 segment that displayed features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic pre-conditioning, and female sex. Thus, our results identified a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both the injury response and protection from ischemic AKI.


Assuntos
Injúria Renal Aguda , Túbulos Renais Proximais , Fator de Transcrição PAX2 , Fator de Transcrição PAX8 , Insuficiência Renal Crônica , Animais , Feminino , Camundongos , Injúria Renal Aguda/complicações , Injúria Renal Aguda/genética , Isquemia/complicações , Túbulos Renais Proximais/patologia , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/genética , Traumatismo por Reperfusão/genética , Fator de Transcrição PAX8/genética , Fator de Transcrição PAX8/metabolismo , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo
2.
Am J Physiol Renal Physiol ; 320(3): F359-F374, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33427061

RESUMO

Lysophosphatidic acid (LPA) increases platelet-derived growth factor-B (PDGFB) and connective tissue growth factor (CTGF) production and secretion by proximal tubule (PT) cells through LPA2 receptor-Gqα-αvß6-integrin-mediated activation of transforming growth factor-ß1 (TGFB1). LPA2, ß6-integrin, PDGFB, and CTGF increase in kidneys after ischemia-reperfusion injury (IRI), coinciding with fibrosis. The TGFB1 receptor antagonist SD-208 prevents increases of ß6-integrin, TGFB1-SMAD signaling, and PDGFB/CTGF expression after IRI and ameliorates fibrosis (Geng H, Lan R, Singha PK, Gilchrist A, Weinreb PH, Violette SM, Weinberg JM, Saikumar P, Venkatachalam MA. Am J Pathol 181: 1236-1249, 2012; Geng H, Lan R, Wang G, Siddiqi AR, Naski MC, Brooks AI, Barnes JL, Saikumar P, Weinberg JM, Venkatachalam MA. Am J Pathol 174: 1291-1308, 2009). We report now that LPA1 receptor signaling through epidermal growth factor receptor (EGFR)-ERK1/2-activator protein-1 cooperates with LPA2-dependent TGFB1 signaling to additively increase PDGFB/CTGF production and secretion by PT cells. Conversely, inhibition of both pathways results in greater suppression of PDGFB/CTGF production and secretion and promotes greater PT cellular differentiation than inhibiting one pathway alone. Antagonism of the LPA-generating enzyme autotaxin suppressed signaling through both pathways. After IRI, kidneys showed not only more LPA2, nuclear SMAD2/3, and PDGFB/CTGF but also increased LPA1 and autotaxin proteins, together with enhanced EGFR/ERK1/2 activation. Remarkably, the TGFB1 receptor antagonist SD-208 prevented all of these abnormalities excepting increased LPA2. SD-208 inhibits only one arm of LPA signaling: LPA2-Gqα-αvß6-integrin-dependent production of active TGFB1 and its receptor-bound downstream effects. Consequently, far-reaching protection by SD-208 against IRI-induced signaling alterations and tubule-interstitial pathology is not fully explained by our data. TGFB1-dependent feedforward modulation of LPA1 signaling is one possibility. SD-208 effects may also involve mitigation of injury caused by IRI-induced TGFB1 signaling in endothelial cells and monocytes. Our results have translational implications for using TGFB1 receptor antagonists, LPA1 and LPA2 inhibitors concurrently, and autotaxin inhibitors in acute kidney injury to prevent the development of chronic kidney disease.


Assuntos
Injúria Renal Aguda/metabolismo , Citocinas/metabolismo , Túbulos Renais Proximais/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Traumatismo por Reperfusão/metabolismo , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Linhagem Celular , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Modelos Animais de Doenças , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrose , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Túbulos Renais Proximais/patologia , Linfocinas/metabolismo , Masculino , Camundongos , Fosforilação , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ratos Sprague-Dawley , Receptores de Ácidos Lisofosfatídicos/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
3.
Cell Mol Life Sci ; 73(11-12): 2285-308, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27066896

RESUMO

The cytoprotective effects of glycine against cell death have been recognized for over 28 years. They are expressed in multiple cell types and injury settings that lead to necrosis, but are still not widely appreciated or considered in the conceptualization of cell death pathways. In this paper, we review the available data on the expression of this phenomenon, its relationship to major pathophysiologic pathways that lead to cell death and immunomodulatory effects, the hypothesis that it involves suppression by glycine of the development of a hydrophilic death channel of molecular dimensions in the plasma membrane, and evidence for its impact on disease processes in vivo.


Assuntos
Morte Celular/fisiologia , Membrana Celular/fisiologia , Citoproteção/fisiologia , Glicina/metabolismo , Animais , Glicina/sangue , Humanos
4.
BMC Nephrol ; 18(1): 130, 2017 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-28395656

RESUMO

BACKGROUND: Ischemia and reperfusion (I/R) is one of the major causes of acute kidney injury (AKI). Citrate reduces hypoxia-induced mitochondrial energetic deficits in isolated proximal tubules. Moreover, citrate anticoagulation is now frequently used in renal replacement therapy. In the present study a rat model of I/R-induced AKI was utilized to examine renal protection by citrate in vivo. METHODS: AKI was induced by bilateral renal clamping (40 min) followed by reperfusion (3 h). Citrate was infused at three different concentrations (0.3 mmol/kg/h; 0.6 mmol/kg/h and 1.0 mmol/kg/h) continuously for 60 min before and 45 min after ischemia. Plasma calcium concentrations were kept stable by infusion of calcium gluconate. The effect of citrate was evaluated by biomonitoring, blood and plasma parameters, histopathology and tissue ATP content. RESULTS: In comparison to the normoxic control group bilateral renal ischemia led to an increase of creatinine and lactate dehydrogenase activity and a decrease in tissue ATP content and was accompanied by a drop in mean arterial blood pressure. Infusion of 1.0 mmol/kg/h citrate led to lower creatinine and reduced LDH activity compared to the I/R control group and a tendency for higher tissue ATP content. Pre-ischemic infusion of 1.0 mmol/kg/h citrate stabilized blood pressure during ischemia. CONCLUSIONS: Citrate has a protective effect during I/R-induced AKI, possibly by limiting the mitochondrial deficit as well as by beneficial cardiovascular effects. This strengthens the rationale of using citrate in continuous renal replacement therapy and encourages consideration of citrate infusion as a therapeutic treatment for AKI in humans.


Assuntos
Injúria Renal Aguda/etiologia , Anticoagulantes/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Ácido Cítrico/farmacologia , Rim/efeitos dos fármacos , Traumatismo por Reperfusão/complicações , Injúria Renal Aguda/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Gluconato de Cálcio/farmacologia , Creatinina/metabolismo , L-Lactato Desidrogenase/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Masculino , Ratos , Artéria Renal , Traumatismo por Reperfusão/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(47): 16836-41, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25385600

RESUMO

Receptor-interacting protein kinase 3 (RIPK3)-mediated necroptosis is thought to be the pathophysiologically predominant pathway that leads to regulated necrosis of parenchymal cells in ischemia-reperfusion injury (IRI), and loss of either Fas-associated protein with death domain (FADD) or caspase-8 is known to sensitize tissues to undergo spontaneous necroptosis. Here, we demonstrate that renal tubules do not undergo sensitization to necroptosis upon genetic ablation of either FADD or caspase-8 and that the RIPK1 inhibitor necrostatin-1 (Nec-1) does not protect freshly isolated tubules from hypoxic injury. In contrast, iron-dependent ferroptosis directly causes synchronized necrosis of renal tubules, as demonstrated by intravital microscopy in models of IRI and oxalate crystal-induced acute kidney injury. To suppress ferroptosis in vivo, we generated a novel third-generation ferrostatin (termed 16-86), which we demonstrate to be more stable, to metabolism and plasma, and more potent, compared with the first-in-class compound ferrostatin-1 (Fer-1). Even in conditions with extraordinarily severe IRI, 16-86 exerts strong protection to an extent which has not previously allowed survival in any murine setting. In addition, 16-86 further potentiates the strong protective effect on IRI mediated by combination therapy with necrostatins and compounds that inhibit mitochondrial permeability transition. Renal tubules thus represent a tissue that is not sensitized to necroptosis by loss of FADD or caspase-8. Finally, ferroptosis mediates postischemic and toxic renal necrosis, which may be therapeutically targeted by ferrostatins and by combination therapy.


Assuntos
Apoptose , Túbulos Renais/citologia , Animais , Peso Corporal , Caspase 8/genética , Caspase 8/fisiologia , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/fisiologia , Camundongos , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia , Traumatismo por Reperfusão/prevenção & controle
6.
J Am Soc Nephrol ; 27(11): 3356-3367, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27000065

RESUMO

During recovery by regeneration after AKI, proximal tubule cells can fail to redifferentiate, undergo premature growth arrest, and become atrophic. The atrophic tubules display pathologically persistent signaling increases that trigger production of profibrotic peptides, proliferation of interstitial fibroblasts, and fibrosis. We studied proximal tubules after ischemia-reperfusion injury (IRI) to characterize possible mitochondrial pathologies and alterations of critical enzymes that govern energy metabolism. In rat kidneys, tubules undergoing atrophy late after IRI but not normally recovering tubules showed greatly reduced mitochondrial number, with rounded profiles, and large autophagolysosomes. Studies after IRI of kidneys in mice, done in parallel, showed large scale loss of the oxidant-sensitive mitochondrial protein Mpv17L. Renal expression of hypoxia markers also increased after IRI. During early and late reperfusion after IRI, kidneys exhibited increased lactate and pyruvate content and hexokinase activity, which are indicators of glycolysis. Furthermore, normally regenerating tubules as well as tubules undergoing atrophy exhibited increased glycolytic enzyme expression and inhibitory phosphorylation of pyruvate dehydrogenase. TGF-ß antagonism prevented these effects. Our data show that the metabolic switch occurred early during regeneration after injury and was reversed during normal tubule recovery but persisted and became progressively more severe in tubule cells that failed to redifferentiate. In conclusion, irreversibility of the metabolic switch, taking place in the context of hypoxia, high TGF-ß signaling and depletion of mitochondria characterizes the development of atrophy in proximal tubule cells and may contribute to the renal pathology after AKI.


Assuntos
Injúria Renal Aguda/complicações , Glicólise , Isquemia/complicações , Túbulos Renais Proximais/patologia , Rim/irrigação sanguínea , Mitocôndrias/metabolismo , Doenças Mitocondriais/etiologia , Animais , Atrofia/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley
7.
BJU Int ; 117(5): 766-74, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26053785

RESUMO

OBJECTIVE: To assess the 1-year renal functional changes in patients undergoing partial nephrectomy with intra-operative renal biopsies. PATIENTS AND METHODS: A total of 40 patients with a single renal mass deemed fit for a partial nephrectomy were recruited prospectively between January 2009 and October 2010. We performed renal biopsies of normal renal parenchyma and collected serum markers before, during and after surgically induced renal clamp ischaemia during the partial nephrectomy. We then followed patients clinically with interval serum creatinine and physical examination. RESULTS: Peri-operative data from 40 patients showed a transient increase in creatinine levels which did not correlate with ischaemia time. Renal ultrastructural changes were generally mild and included mitochondrial swelling, which resolved at the post-perfusion biopsy. A total of 37 patients had 1-year follow-up data. Creatinine at 1 year increased by 0.121 mg/dL, which represents a 12.99% decrease in renal function from baseline (preoperative creatinine 0.823 mg/dL, estimated glomerular filtration rate = 93.9 mL/min/1.73 m(2) ). The only factors predicting creatinine change on multivariate analysis were patient age, race and ischaemia type, with cold ischaemia being associated with higher creatinine level. Importantly, the duration of ischaemia did not show any significant correlation with renal function change, either as a continuous variable (P = 0.452) or as a categorical variable (P = 0.792). CONCLUSIONS: Our data suggest that limited ischaemia is generally well tolerated in the setting of partial nephrectomy and does not directly correspond to long-term renal functional decline. For surgeons performing partial nephrectomy, the kidney can be safely clamped to ensure optimum oncological outcomes.


Assuntos
Isquemia Fria , Neoplasias Renais/cirurgia , Rim/fisiologia , Nefrectomia/métodos , Isquemia Quente , Adulto , Idoso , Idoso de 80 Anos ou mais , Isquemia Fria/efeitos adversos , Constrição , Creatinina/sangue , Progressão da Doença , Feminino , Seguimentos , Taxa de Filtração Glomerular , Humanos , Rim/irrigação sanguínea , Rim/ultraestrutura , Neoplasias Renais/fisiopatologia , Masculino , Pessoa de Meia-Idade , Nefrectomia/efeitos adversos , Estudos Prospectivos , Artéria Renal , Insuficiência Renal Crônica/classificação , Insuficiência Renal Crônica/etiologia , Fatores de Tempo , Isquemia Quente/efeitos adversos
8.
Proc Natl Acad Sci U S A ; 110(29): 12024-9, 2013 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-23818611

RESUMO

Regulated necrosis (RN) may result from cyclophilin (Cyp)D-mediated mitochondrial permeability transition (MPT) and receptor-interacting protein kinase (RIPK)1-mediated necroptosis, but it is currently unclear whether there is one common pathway in which CypD and RIPK1 act in or whether separate RN pathways exist. Here, we demonstrate that necroptosis in ischemia-reperfusion injury (IRI) in mice occurs as primary organ damage, independent of the immune system, and that mice deficient for RIPK3, the essential downstream partner of RIPK1 in necroptosis, are protected from IRI. Protection of RIPK3-knockout mice was significantly stronger than of CypD-deficient mice. Mechanistically, in vivo analysis of cisplatin-induced acute kidney injury and hyperacute TNF-shock models in mice suggested the distinctness of CypD-mediated MPT from RIPK1/RIPK3-mediated necroptosis. We, therefore, generated CypD-RIPK3 double-deficient mice that are viable and fertile without an overt phenotype and that survived prolonged IRI, which was lethal to each single knockout. Combined application of the RIPK1 inhibitor necrostatin-1 and the MPT inhibitor sanglifehrin A confirmed the results with mutant mice. The data demonstrate the pathophysiological coexistence and corelevance of two separate pathways of RN in IRI and suggest that combination therapy targeting distinct RN pathways can be beneficial in the treatment of ischemic injury.


Assuntos
Apoptose/fisiologia , Ciclofilinas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Necrose/fisiopatologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Traumatismo por Reperfusão/complicações , Animais , Linhagem Celular , Peptidil-Prolil Isomerase F , Ciclofilinas/genética , Primers do DNA/genética , Genótipo , Estimativa de Kaplan-Meier , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poro de Transição de Permeabilidade Mitocondrial , Necrose/etiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética
9.
J Am Soc Nephrol ; 26(8): 1765-76, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25810494

RESUMO

The transition of AKI to CKD has major clinical significance. As reviewed here, recent studies show that a subpopulation of dedifferentiated, proliferating tubules recovering from AKI undergo pathologic growth arrest, fail to redifferentiate, and become atrophic. These abnormal tubules exhibit persistent, unregulated, and progressively increasing profibrotic signaling along multiple pathways. Paracrine products derived therefrom perturb normal interactions between peritubular capillary endothelium and pericyte-like fibroblasts, leading to myofibroblast transformation, proliferation, and fibrosis as well as capillary disintegration and rarefaction. Although signals from injured endothelium and inflammatory/immune cells also contribute, tubule injury alone is sufficient to produce the interstitial pathology required for fibrosis. Localized hypoxia produced by microvascular pathology may also prevent tubule recovery. However, fibrosis is not intrinsically progressive, and microvascular pathology develops strictly around damaged tubules; thus, additional deterioration of kidney structure after the transition of AKI to CKD requires new acute injury or other mechanisms of progression. Indeed, experiments using an acute-on-chronic injury model suggest that additional loss of parenchyma caused by failed repair of AKI in kidneys with prior renal mass reduction triggers hemodynamically mediated processes that damage glomeruli to cause progression. Continued investigation of these pathologic mechanisms should reveal options for preventing renal disease progression after AKI.


Assuntos
Injúria Renal Aguda/complicações , Túbulos Renais/fisiopatologia , Insuficiência Renal Crônica/etiologia , Injúria Renal Aguda/fisiopatologia , Capilares/fisiopatologia , Progressão da Doença , Humanos , Hipóxia/complicações , Túbulos Renais/metabolismo , Nefroesclerose/etiologia , Nefroesclerose/metabolismo , Circulação Renal , Vasoconstrição
10.
Kidney Int ; 88(3): 434-7, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26323068

RESUMO

A report by Neelisetty et al. suggests that TGFBR2 deletion from matrix-producing interstitial cells results in decreased transforming growth factor-ß (TGF-ß) signaling in the cells but does not decrease renal fibrosis after injury. Considered in the context of TGF-ß signaling in different cell types involved in renal fibrosis and the existence of other ligands that may produce fibrosis, these findings are provocative, but owing to technical issues of recombination efficiency in inducible models of Cre-lox gene deletion, further studies are needed.


Assuntos
Matriz Extracelular/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais
11.
J Am Chem Soc ; 136(12): 4551-6, 2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24592866

RESUMO

Ferrostatin-1 (Fer-1) inhibits ferroptosis, a form of regulated, oxidative, nonapoptotic cell death. We found that Fer-1 inhibited cell death in cellular models of Huntington's disease (HD), periventricular leukomalacia (PVL), and kidney dysfunction; Fer-1 inhibited lipid peroxidation, but not mitochondrial reactive oxygen species formation or lysosomal membrane permeability. We developed a mechanistic model to explain the activity of Fer-1, which guided the development of ferrostatins with improved properties. These studies suggest numerous therapeutic uses for ferrostatins, and that lipid peroxidation mediates diverse disease phenotypes.


Assuntos
Cicloexilaminas/farmacologia , Doença de Huntington/tratamento farmacológico , Nefropatias/tratamento farmacológico , Leucomalácia Periventricular/tratamento farmacológico , Lipídeos de Membrana/metabolismo , Fenilenodiaminas/farmacologia , Morte Celular/efeitos dos fármacos , Cicloexilaminas/uso terapêutico , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Leucomalácia Periventricular/metabolismo , Leucomalácia Periventricular/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Fenilenodiaminas/uso terapêutico
12.
J Am Soc Nephrol ; 24(3): 506-17, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23411786

RESUMO

Tolerance of the human kidney to ischemia is controversial. Here, we prospectively studied the renal response to clamp ischemia and reperfusion in humans, including changes in putative biomarkers of AKI. We performed renal biopsies before, during, and after surgically induced renal clamp ischemia in 40 patients undergoing partial nephrectomy. Ischemia duration was >30 minutes in 82.5% of patients. There was a mild, transient increase in serum creatinine, but serum cystatin C remained stable. Renal functional changes did not correlate with ischemia duration. Renal structural changes were much less severe than observed in animal models that used similar durations of ischemia. Other biomarkers were only mildly elevated and did not correlate with renal function or ischemia duration. In summary, these data suggest that human kidneys can safely tolerate 30-60 minutes of controlled clamp ischemia with only mild structural changes and no acute functional loss.


Assuntos
Isquemia/fisiopatologia , Rim/irrigação sanguínea , Rim/fisiopatologia , Nefrectomia/métodos , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Injúria Renal Aguda/prevenção & controle , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Creatinina/sangue , Cistatina C/sangue , Feminino , Humanos , Isquemia/patologia , Rim/patologia , Masculino , Pessoa de Meia-Idade , Nefrectomia/efeitos adversos , Estudos Prospectivos , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Traumatismo por Reperfusão/prevenção & controle , Fatores de Tempo
13.
J Am Soc Nephrol ; 24(10): 1545-57, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23833261

RESUMO

The pathophysiology of contrast-induced AKI (CIAKI) is incompletely understood due to the lack of an appropriate in vivo model that demonstrates reduced kidney function before administration of radiocontrast media (RCM). Here, we examine the effects of CIAKI in vitro and introduce a murine ischemia/reperfusion injury (IRI)-based approach that allows induction of CIAKI by a single intravenous application of standard RCM after injury for in vivo studies. Whereas murine renal tubular cells and freshly isolated renal tubules rapidly absorbed RCM, plasma membrane integrity and cell viability remained preserved in vitro and ex vivo, indicating that RCM do not induce apoptosis or regulated necrosis of renal tubular cells. In vivo, the IRI-based CIAKI model exhibited typical features of clinical CIAKI, including RCM-induced osmotic nephrosis and increased serum levels of urea and creatinine that were not altered by inhibition of apoptosis. Direct evaluation of renal morphology by intravital microscopy revealed dilation of renal tubules and peritubular capillaries within 20 minutes of RCM application in uninjured mice and similar, but less dramatic, responses after IRI pretreatment. Necrostatin-1 (Nec-1), a specific inhibitor of the receptor-interacting protein 1 (RIP1) kinase domain, prevented osmotic nephrosis and CIAKI, whereas an inactive Nec-1 derivate (Nec-1i) or the pan-caspase inhibitor zVAD did not. In addition, Nec-1 prevented RCM-induced dilation of peritubular capillaries, suggesting a novel role unrelated to cell death for the RIP1 kinase domain in the regulation of microvascular hemodynamics and pathophysiology of CIAKI.


Assuntos
Injúria Renal Aguda/prevenção & controle , Imidazóis/uso terapêutico , Indóis/uso terapêutico , Túbulos Renais/efeitos dos fármacos , Nefrose/prevenção & controle , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Injúria Renal Aguda/induzido quimicamente , Animais , Morte Celular , Linhagem Celular , Meios de Contraste/toxicidade , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Imidazóis/farmacologia , Indóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nefrose/induzido quimicamente , Traumatismo por Reperfusão/complicações
14.
Kidney Int ; 84(1): 11-4, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23812359

RESUMO

Shen et al. exploit glycobiology to dampen transforming growth factor-ß (TGF-ß) signaling and ameliorate renal fibrosis after ureteral obstruction. Core fucosylation of N-linked oligosaccharides in TGF-ß receptors is required for receptor function. Adenoviruses expressing Fut8-fucosyl transferase-shRNA inhibited receptor fucosylation, decreased tubule TGF-ß signaling, and reduced fibrosis. Fut8-shRNA interferes with core fucosylation of other receptors also. Regardless, this first attempt to capitalize on a new aspect of TGF-ß receptor function provides a basis for further research.


Assuntos
Fucose/metabolismo , Fucosiltransferases/metabolismo , Nefropatias/prevenção & controle , Rim/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Humanos , Masculino
15.
Kidney Int ; 84(1): 16-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23812361

RESUMO

Kim et al. show that isoflurane uses a tubule-based transforming growth factor-ß/CD73-dependent process that generates adenosine to protect mice from ischemic acute kidney injury (AKI) with effects to prevent the 'no-reflow phenomenon' and decrease inflammation. While direct cytoprotection occurred in culture, extensive research suggests that in vivo adenosine protection from rodent ischemic AKI is mediated by a mutually cooperative mechanism involving blood flow, inflammation, and innate immunity through multiple adenosine receptors with promiscuous actions on diverse cell types.


Assuntos
5'-Nucleotidase/biossíntese , Injúria Renal Aguda/prevenção & controle , Anestésicos Inalatórios/farmacologia , Isoflurano/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Animais , Proteínas Ligadas por GPI/biossíntese , Humanos , Masculino
16.
Am J Pathol ; 181(4): 1236-49, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22885106

RESUMO

After ischemia-reperfusion injury (IRI), kidney tubules show activated transforming growth factor ß (TGF-ß) signaling and increased expression of profibrotic peptides, platelet-derived growth factor-B (PDGF-B) and connective tissue growth factor (CTGF). If tubule repair after IRI is incomplete, sustained paracrine activity of these peptides can activate interstitial fibroblast progenitors and cause fibrosis. We show that lysophosphatidic acid (LPA), a ubiquitous phospholipid that is increased at sites of injury and inflammation, signals through LPA2 receptors and Gαq proteins of cultured proximal tubule cells to transactivate latent TGF-ß in a Rho/Rho-kinase and αvß6 integrin-dependent manner. Active TGF-ß peptide then initiates signaling to increase the production and secretion of PDGF-B and CTGF. In a rat model of IRI, increased TGF-ß signaling that was initiated early during reperfusion did not subside during recovery, but progressively increased, causing tubulointerstitial fibrosis. This was accompanied by correspondingly increased LPA2 and ß6 integrin proteins and elevated tubule expression of TGF-ß1, together with PDGF-B and CTGF. Treatment with a pharmacological TGF-ß type I receptor antagonist suppressed TGF-ß signaling, decreased the expression of ß6 integrin, PDGF-B, and CTGF, and ameliorated fibrosis. We suggest that LPA-initiated autocrine signaling is a potentially important mechanism that gives rise to paracrine profibrotic signaling in injured kidney tubule cells.


Assuntos
Antígenos de Neoplasias/metabolismo , Citocinas/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Integrinas/metabolismo , Túbulos Renais Proximais/metabolismo , Lisofosfolipídeos/farmacologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Citocinas/genética , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Lipídeos/sangue , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo
18.
bioRxiv ; 2023 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-37873377

RESUMO

Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. In this report, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI. We found that Pax2 and Pax8 are upregulated after severe AKI and correlate with chronic injury. Surprisingly, we then discovered that proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to preconditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of S3 proximal tubule cells that display features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic preconditioning, and female sex. Taken together, our results identify a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both injury response and protection from ischemic AKI. TRANSLATIONAL STATEMENT: Identifying the molecular and genetic regulators unique to the nephron that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are two homologous nephron-specific transcription factors that are critical for kidney development and physiology. Here we report that proximal-tubule-selective depletion of Pax2 and Pax8 protects against both acute and chronic injury and induces an expression profile in the S3 proximal tubule with common features shared among diverse conditions that protect against ischemia. These findings highlight a new role for Pax proteins as potential therapeutic targets to treat AKI.

19.
Am J Physiol Renal Physiol ; 303(1): F56-63, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22513847

RESUMO

Acute kidney injury (AKI) is the most common kidney disease in hospitalized patients with high mortality. Ischemia and reperfusion (I/R) is one of the major causes of AKI. The combination of α-ketoglutarate+malate (αKG/MAL) showed the ability to reduce hypoxia-induced damage to isolated proximal tubules. The present study utilizes a rat model of I/R-induced AKI accompanied by intensive biomonitoring to examine whether αKG/MAL provides protection in vivo. AKI was induced in male Sprague-Dawley rats by bilateral renal clamping (40 min) followed by reperfusion (240 min). αKG/MAL was infused continuously for 60 min before and 45 min after ischemia. Normoxic and I/R control groups received 0.9% NaCl solution. The effect of αKG/MAL was evaluated by biomonitoring, blood and plasma parameters, histopathology, and immunohistochemical staining for kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL), as well as by determination of tissue ATP and nonesterified fatty acid concentrations. Intravenous infusion of αKG/MAL at a cumulative dose of 1 mmol/kg each (146 mg/kg αKG and 134 mg/kg MAL) did not prevent I/R-induced increases in plasma creatinine, histopathological alterations, or cortical ATP depletion. On the contrary, the most notable adverse affect in animals receiving αKG/MAL was the decrease in mean arterial blood pressure, which was also accompanied by a reduction in heart rate. Supplementation with αKG/MAL, which is very protective against hypoxia-induced injury in isolated proximal tubules, does not protect against I/R-induced renal injury in vivo, possibly due to cardiovascular depressive effects.


Assuntos
Injúria Renal Aguda/patologia , Pressão Sanguínea/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Ácidos Cetoglutáricos/toxicidade , Rim/efeitos dos fármacos , Malatos/toxicidade , Traumatismo por Reperfusão/patologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Animais , Moléculas de Adesão Celular/metabolismo , Modelos Animais de Doenças , Hipóxia/metabolismo , Hipóxia/patologia , Hipóxia/fisiopatologia , Ácidos Cetoglutáricos/farmacologia , Ácidos Cetoglutáricos/uso terapêutico , Rim/irrigação sanguínea , Rim/metabolismo , Rim/patologia , Malatos/farmacologia , Malatos/uso terapêutico , Masculino , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle
20.
Am J Physiol Renal Physiol ; 302(9): F1210-23, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22301622

RESUMO

We investigated the signaling basis for tubule pathology during fibrosis after renal injury. Numerous signaling pathways are activated physiologically to direct tubule regeneration after acute kidney injury (AKI) but several persist pathologically after repair. Among these, transforming growth factor (TGF)-ß is particularly important because it controls epithelial differentiation and profibrotic cytokine production. We found that increased TGF-ß signaling after AKI is accompanied by PTEN loss from proximal tubules (PT). With time, subpopulations of regenerating PT with persistent loss of PTEN (phosphate and tension homolog) failed to differentiate, became growth arrested, expressed vimentin, displayed profibrotic JNK activation, and produced PDGF-B. These tubules were surrounded by fibrosis. In contrast, PTEN recovery was associated with epithelial differentiation, normal tubule repair, and less fibrosis. This beneficial outcome was promoted by TGF-ß antagonism. Tubule-specific induction of TGF-ß led to PTEN loss, JNK activation, and fibrosis even without prior AKI. In PT culture, high TGF-ß depleted PTEN, inhibited differentiation, and activated JNK. Conversely, TGF-ß antagonism increased PTEN, promoted differentiation, and decreased JNK activity. Cre-Lox PTEN deletion suppressed differentiation, induced growth arrest, and activated JNK. The low-PTEN state with JNK signaling and fibrosis was ameliorated by contralateral nephrectomy done 2 wk after unilateral ischemia, suggesting reversibility of the low-PTEN dysfunctional tubule phenotype. Vimentin-expressing tubules with low-PTEN and JNK activation were associated with fibrosis also after tubule-selective AKI, and with human chronic kidney diseases of diverse etiology. By preventing tubule differentiation, the low-PTEN state may provide a platform for signals initiated physiologically to persist pathologically and cause fibrosis after injury.


Assuntos
Diferenciação Celular , Túbulos Renais Proximais/patologia , MAP Quinase Quinase 4/fisiologia , PTEN Fosfo-Hidrolase/deficiência , Fenótipo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Células Cultivadas , Doença Crônica , Fibrose , Humanos , Nefropatias/patologia , Nefropatias/fisiopatologia , Túbulos Renais Proximais/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Ratos , Ratos Sprague-Dawley , Regeneração/fisiologia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA