Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 183(2): 474-489.e17, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33035451

RESUMO

Mg2+ is the most abundant divalent cation in metazoans and an essential cofactor for ATP, nucleic acids, and countless metabolic enzymes. To understand how the spatio-temporal dynamics of intracellular Mg2+ (iMg2+) are integrated into cellular signaling, we implemented a comprehensive screen to discover regulators of iMg2+ dynamics. Lactate emerged as an activator of rapid release of Mg2+ from endoplasmic reticulum (ER) stores, which facilitates mitochondrial Mg2+ (mMg2+) uptake in multiple cell types. We demonstrate that this process is remarkably temperature sensitive and mediated through intracellular but not extracellular signals. The ER-mitochondrial Mg2+ dynamics is selectively stimulated by L-lactate. Further, we show that lactate-mediated mMg2+ entry is facilitated by Mrs2, and point mutations in the intermembrane space loop limits mMg2+ uptake. Intriguingly, suppression of mMg2+ surge alleviates inflammation-induced multi-organ failure. Together, these findings reveal that lactate mobilizes iMg2+ and links the mMg2+ transport machinery with major metabolic feedback circuits and mitochondrial bioenergetics.


Assuntos
Retículo Endoplasmático/metabolismo , Ácido Láctico/metabolismo , Magnésio/metabolismo , Animais , Células COS , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Chlorocebus aethiops , Retículo Endoplasmático/fisiologia , Feminino , Células HeLa , Células Hep G2 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo
2.
Sci Signal ; 13(628)2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32317369

RESUMO

The tricarboxylic acid (TCA) cycle converts the end products of glycolysis and fatty acid ß-oxidation into the reducing equivalents NADH and FADH2 Although mitochondrial matrix uptake of Ca2+ enhances ATP production, it remains unclear whether deprivation of mitochondrial TCA substrates alters mitochondrial Ca2+ flux. We investigated the effect of TCA cycle substrates on MCU-mediated mitochondrial matrix uptake of Ca2+, mitochondrial bioenergetics, and autophagic flux. Inhibition of glycolysis, mitochondrial pyruvate transport, or mitochondrial fatty acid transport triggered expression of the MCU gatekeeper MICU1 but not the MCU core subunit. Knockdown of mitochondrial pyruvate carrier (MPC) isoforms or expression of the dominant negative mutant MPC1R97W resulted in increased MICU1 protein abundance and inhibition of MCU-mediated mitochondrial matrix uptake of Ca2+ We also found that genetic ablation of MPC1 in hepatocytes and mouse embryonic fibroblasts resulted in reduced resting matrix Ca2+, likely because of increased MICU1 expression, but resulted in changes in mitochondrial morphology. TCA cycle substrate-dependent MICU1 expression was mediated by the transcription factor early growth response 1 (EGR1). Blocking mitochondrial pyruvate or fatty acid flux was linked to increased autophagy marker abundance. These studies reveal a mechanism that controls the MCU-mediated Ca2+ flux machinery and that depends on TCA cycle substrate availability. This mechanism generates a metabolic homeostatic circuit that protects cells from bioenergetic crisis and mitochondrial Ca2+ overload during periods of nutrient stress.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Ácidos Graxos/metabolismo , Mitocôndrias Hepáticas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas Mitocondriais/metabolismo , Ácido Pirúvico/metabolismo , Animais , Transporte Biológico Ativo/genética , Canais de Cálcio/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Transporte de Cátions/genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Camundongos Knockout , Mitocôndrias Hepáticas/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas Mitocondriais/genética
3.
Trans Am Clin Climatol Assoc ; 130: 33-40, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31516162

RESUMO

Acute kidney injury (AKI) is common among hospitalized patients and is associated with high morbidity and mortality. Inflammation is recognized to play an important role in both ischemic and toxic models of AKI. Cisplatin is a widely used and highly effective cancer chemotherapeutic agent but carries the risk of nephrotoxicity. We have used a model of cisplatin-induced AKI to explore the functions of the innate immune response in kidney injury. Several components of innate immunity, such as Toll-like receptor sensing and inflammatory cytokine production, contribute to both ischemic and cisplatin-induced AKI. Importantly, it is the activity of these components in kidney parenchymal cells, rather than immune cells, which mediate AKI. Cellular components of innate immunity, such as neutrophils and dendritic cells, appear to play disparate roles in ischemic vs toxic AKI. Innate immune pathways could be targeted to prevent or treat AKI.


Assuntos
Injúria Renal Aguda/imunologia , Células Dendríticas/imunologia , Imunidade Inata/imunologia , Inflamação/imunologia , Neutrófilos/imunologia , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/imunologia , Injúria Renal Aguda/induzido quimicamente , Animais , Antineoplásicos/toxicidade , Cisplatino/toxicidade , Modelos Animais de Doenças , Rim/imunologia , Camundongos
4.
J Am Assoc Lab Anim Sci ; 58(1): 21-29, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30538007

RESUMO

Urinary biomarkers are used increasingly for sensitive prediction of kidney injury in preclinical and clinical studies. Given the frequent requirement of anesthesia in various animal models of disease, it is important to define the effects of anesthesia on kidney injury biomarkers to guide the appropriate selection of anesthetic agents and to avoid potential confounders in the interpretation of data. Therefore, we performed a prospective study using male C57BL/6J mice (n = 45) exposed to a single anesthetic episode to determine the effects several common anesthesia regimens on the urinary excretion of 2 commonly used kidney injury biomarkers: hepatitis A virus cellular receptor 1 (HAVCR1, also known as KIM1) and lipocalin 2 (LCN2, also known as NGAL). We evaluated 3 injectable regimens (ketamine-xylazine, tiletamine-zolazepam, and pentobarbital) and 2 inhalational agents (isoflurane and sevoflurane). Concentrations of HAVCR1 and LCN2 in urine collected at various time points after anesthesia were measured by using ELISA. Administration of ketamine-xylazine resulted in a significant increase in HAVCR1 levels at 6 h after anesthesia but a decrease in LCN2 levels compared with baseline. LCN2 levels steadily increased over the first 24 h after inhalant anesthesia, with a significant increase at 24 h after sevoflurane. These results suggest that injectable anesthesia had early effects on HAVCR1 and LCN2 levels, whereas inhalational agents increased these biomarkers over prolonged time.


Assuntos
Anestesia Geral/veterinária , Anestésicos/administração & dosagem , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Lipocalina-2/urina , Anestesia Geral/efeitos adversos , Anestésicos/efeitos adversos , Animais , Biomarcadores/urina , Regulação da Expressão Gênica/efeitos dos fármacos , Rim/efeitos dos fármacos , Ciência dos Animais de Laboratório , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estudos Prospectivos
5.
Am J Physiol Renal Physiol ; 313(2): F522-F534, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28515179

RESUMO

Novel therapeutic interventions for preventing or attenuating kidney injury following ischemia-reperfusion injury (IRI) remain a focus of significant interest. Currently, there are no definitive therapeutic or preventive approaches available for ischemic acute kidney injury (AKI). Our objective is to determine 1) whether renal arginase activity or expression is increased in renal IRI, and 2) whether arginase plays a role in development of renal IRI. The impact of arginase activity and expression on renal damage was evaluated in male C57BL/6J (wild type) and arginase-2 (ARG2)-deficient (Arg2-/- ) mice subjected to bilateral renal ischemia for 28 min, followed by reperfusion for 24 h. ARG2 expression and arginase activity significantly increased following renal IRI, paralleling the increase in kidney injury. Pharmacological blockade or genetic deficiency of Arg2 conferred kidney protection in renal IRI. Arg2-/- mice had significantly attenuated kidney injury and lower plasma creatinine and blood urea nitrogen levels after renal IRI. Blocking arginases using S-(2-boronoethyl)-l-cysteine (BEC) 18 h before ischemia mimicked arginase deficiency by reducing kidney injury, histopathological changes and kidney injury marker-1 expression, renal apoptosis, kidney inflammatory cell recruitment and inflammatory cytokines, and kidney oxidative stress; increasing kidney nitric oxide (NO) production and endothelial NO synthase (eNOS) phosphorylation, kidney peroxisome proliferator-activated receptor-γ coactivator-1α expression, and mitochondrial ATP; and preserving kidney mitochondrial ultrastructure compared with vehicle-treated IRI mice. Importantly, BEC-treated eNOS-knockout mice failed to reduce blood urea nitrogen and creatinine following renal IRI. These findings indicate that ARG2 plays a major role in renal IRI, via an eNOS-dependent mechanism, and that blocking ARG2 activity or expression could be a novel therapeutic approach for prevention of AKI.


Assuntos
Injúria Renal Aguda/enzimologia , Arginase/metabolismo , Traumatismo por Reperfusão/enzimologia , Injúria Renal Aguda/patologia , Trifosfato de Adenosina/metabolismo , Animais , Arginase/antagonistas & inibidores , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Nitritos/metabolismo , Estresse Oxidativo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Traumatismo por Reperfusão/patologia
6.
Kidney Int ; 91(3): 671-682, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27914709

RESUMO

Inflammation is a central pathophysiologic mechanism that contributes to diabetes mellitus and diabetic nephropathy. Recently, we showed that macrophages directly contribute to diabetic renal injury and that pharmacological blockade or genetic deficiency of chemokine (C-C motif) receptor 2 (CCR2) confers kidney protection in diabetic nephropathy. However, the direct role of CCR2 in kidney-derived cells such as podocytes in diabetic nephropathy remains unclear. To study this, we developed a transgenic mouse model expressing CCR2 specifically in podocytes (Tg[NPHS2-Ccr2]) on a nephropathy-prone (DBA/2J) and CCR2-deficient (Ccr2-/-) background with heterozygous Ccr2+/- littermate controls. Diabetes was induced by streptozotocin. As expected, absence of CCR2 conferred kidney protection after nine weeks of diabetes. In contrast, transgenic CCR2 overexpression in the podocytes of Ccr2-/- mice resulted in significantly increased albuminuria, blood urea nitrogen, histopathologic changes, kidney fibronectin and type 1 collagen expression, podocyte loss, and glomerular apoptosis after nine weeks of streptozotocin-induced diabetes. Interestingly, there was no concurrent increase in kidney macrophage recruitment or inflammatory cytokine levels in the mice. These findings support a direct role for CCR2 expression in podocytes to mediate diabetic renal injury, independent of monocyte/macrophage recruitment. Thus, targeting the CCR2 signaling cascade in podocytes could be a novel therapeutic approach for treatment of diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Nefropatias Diabéticas/metabolismo , Podócitos/metabolismo , Receptores CCR2/metabolismo , Albuminúria/genética , Albuminúria/metabolismo , Albuminúria/prevenção & controle , Animais , Apoptose , Nitrogênio da Ureia Sanguínea , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/prevenção & controle , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrose , Predisposição Genética para Doença , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Monócitos/metabolismo , Fenótipo , Podócitos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CCR2/deficiência , Receptores CCR2/genética , Transdução de Sinais , Estreptozocina , Regulação para Cima
7.
PLoS One ; 11(10): e0164832, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27788160

RESUMO

BACKGROUND: Although stored urine samples are often used in biomarker studies focused on acute and chronic kidney disease, how storage time impacts biomarker levels is not well understood. METHODS: 866 subjects enrolled in the NIDDK-sponsored ASsessment, Serial Evaluation, and Subsequent Sequelae in Acute Kidney Injury (ASSESS-AKI) Study were included. Samples were processed under standard conditions and stored at -70°C until analyzed. Kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), interleukin-18 (IL-18), and liver fatty acid binding protein (L-FABP) were measured in urine samples collected during the index hospitalization or an outpatient visit 3 months later. Mixed effects models were used to determine the effect of storage time on biomarker levels and stratified by visit. RESULTS: Median storage was 17.8 months (25-75% IQR 10.6-23.7) for samples from the index hospitalization and 14.6 months (IQR 7.3-20.4) for outpatient samples. In the mixed effects models, the only significant association between storage time and biomarker concentration was for KIM-1 in outpatient samples, where each month of storage was associated with a 1.7% decrease (95% CI -3% to -0.3%). There was no relationship between storage time and KIM-1 levels in samples from the index hospitalization. CONCLUSION: There was no significant impact of storage time over a median of 18 months on urine KIM-1, NGAL, IL-18 or L-FABP in hospitalized samples; a statistically significant effect towards a decrease over time was noted for KIM-1 in outpatient samples. Additional studies are needed to determine whether longer periods of storage at -70°C systematically impact levels of these analytes.


Assuntos
Coleta de Urina/métodos , Urina/química , Injúria Renal Aguda/urina , Biomarcadores/urina , Proteínas de Ligação a Ácido Graxo/urina , Feminino , Receptor Celular 1 do Vírus da Hepatite A/análise , Humanos , Interleucina-18/urina , Lipocalina-2/urina , Masculino , Pessoa de Meia-Idade , Insuficiência Renal Crônica/urina , Fatores de Tempo , Coleta de Urina/estatística & dados numéricos
8.
Nanoscale ; 8(8): 4613-22, 2016 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-26846189

RESUMO

Determining ultratrace amounts of protein biomarkers in patient samples in a straightforward and quantitative manner is extremely important for early disease diagnosis and treatment. Here, we successfully demonstrate the novel use of zinc oxide nanorods (ZnO NRs) in the ultrasensitive and quantitative detection of two acute kidney injury (AKI)-related protein biomarkers, tumor necrosis factor (TNF)-α and interleukin (IL)-8, directly from patient samples. We first validate the ZnO NRs-based IL-8 results via comparison with those obtained from using a conventional enzyme-linked immunosorbent method in samples from 38 individuals. We further assess the full detection capability of the ZnO NRs-based technique by quantifying TNF-α, whose levels in human urine are often below the detection limits of conventional methods. Using the ZnO NR platforms, we determine the TNF-α concentrations of all 46 patient samples tested, down to the fg per mL level. Subsequently, we screen for TNF-α levels in approximately 50 additional samples collected from different patient groups in order to demonstrate a potential use of the ZnO NRs-based assay in assessing cytokine levels useful for further clinical monitoring. Our research efforts demonstrate that ZnO NRs can be straightforwardly employed in the rapid, ultrasensitive, quantitative, and simultaneous detection of multiple AKI-related biomarkers directly in patient urine samples, providing an unparalleled detection capability beyond those of conventional analysis methods. Additional key advantages of the ZnO NRs-based approach include a fast detection speed, low-volume assay condition, multiplexing ability, and easy automation/integration capability to existing fluorescence instrumentation. Therefore, we anticipate that our ZnO NRs-based detection method will be highly beneficial for overcoming the frequent challenges in early biomarker development and treatment assessment, pertaining to the facile and ultrasensitive quantification of hard-to-trace biomolecules.


Assuntos
Injúria Renal Aguda/diagnóstico , Biomarcadores/urina , Ensaio de Imunoadsorção Enzimática , Nanotubos/química , Óxido de Zinco/química , Humanos , Interleucina-8/urina , Microscopia Eletrônica de Varredura , Nanotubos/ultraestrutura , Fator de Necrose Tumoral alfa/urina
9.
Toxins (Basel) ; 7(8): 3245-56, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26295408

RESUMO

Cisplatin is a very effective chemotherapeutic agent used against a wide range of solid tumors. A major adverse effect of cisplatin therapy is acute kidney injury (AKI). Neutrophils are reported to infiltrate and exacerbate injury in a wide range of sterile inflammatory models of tissue injury. Here, we studied the kinetics of neutrophil infiltration into kidneys and their role in cisplatin-mediated AKI. Mice treated with cisplatin showed an increase in circulating neutrophils 24 and 48 h after cisplatin administration. Cisplatin treatment caused an increase in kidney leukocytes with neutrophils accounting for the majority of the infiltrating leukocytes. The extent of neutrophil infiltration coincided with the severity of kidney injury and renal dysfunction. To examine the functional relevance of infiltrating neutrophils in cisplatin nephrotoxicity, we depleted neutrophils using a neutrophil-specific antibody (anti-Ly-6G). This antibody resulted in greater than 90% depletion of neutrophils in both the blood and kidney. Of note, depletion of neutrophils had no impact on the extent of cisplatin-induced AKI as compared to non-depleted mice. Earlier, we reported that dendritic cell depletion in CD11c-DTRtg mice causes exacerbation of AKI and a dramatic increase in renal neutrophils. Thus, we also examined the role of neutrophils in dendritic cell-depleted mice treated with cisplatin. Dendritic cell depletion exacerbated AKI in spite of neutrophil depletion. These data demonstrate that cisplatin nephrotoxicity is not mediated by neutrophils and that dendritic cells protect kidneys via neutrophil-independent mechanisms.


Assuntos
Injúria Renal Aguda/imunologia , Antineoplásicos/toxicidade , Cisplatino/toxicidade , Células Dendríticas/imunologia , Injúria Renal Aguda/induzido quimicamente , Animais , Citoproteção , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/imunologia
10.
J Clin Invest ; 124(11): 4989-5001, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25295536

RESUMO

Ischemia is a leading cause of acute kidney injury. Kidney ischemia is associated with loss of cellular ion homeostasis; however, the pathways that underlie ion homeostasis dysfunction are poorly understood. Here, we evaluated the nonselective cation channel transient receptor potential melastatin 2 (TRPM2) in a murine model of kidney ischemia/reperfusion (I/R) injury. TRPM2-deficient mice were resistant to ischemic injury, as reflected by improved kidney function, reduced histologic damage, suppression of proapoptotic pathways, and reduced inflammation. Moreover, pharmacologic TRPM2 inhibition was also protective against I/R injury. TRPM2 was localized mainly in kidney proximal tubule epithelial cells, and studies in chimeric mice indicated that the effects of TRPM2 are due to expression in parenchymal cells rather than hematopoietic cells. TRPM2-deficient mice had less oxidative stress and lower levels of NADPH oxidase activity after ischemia. While RAC1 is a component of the NADPH oxidase complex, its relation to TRPM2 and kidney ischemic injury is unknown. Following kidney ischemia, TRPM2 promoted RAC1 activation, with active RAC1 physically interacting with TRPM2 and increasing TRPM2 expression at the cell membrane. Finally, inhibition of RAC1 reduced oxidant stress and ischemic injury in vivo. These results demonstrate that TRPM2-dependent RAC1 activation increases oxidant stress and suggest that therapeutic approaches targeting TRPM2 and/or RAC1 may be effective in reducing ischemic kidney injury.


Assuntos
Injúria Renal Aguda/enzimologia , Isquemia/enzimologia , Rim/irrigação sanguínea , Neuropeptídeos/metabolismo , Estresse Oxidativo , Canais de Cátion TRPM/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Apoptose , Membrana Celular/metabolismo , Ativação Enzimática , Células Epiteliais/fisiologia , Células HEK293 , Humanos , Peróxido de Hidrogênio/metabolismo , Rim/enzimologia , Túbulos Renais Proximais , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transporte Proteico , Traumatismo por Reperfusão/enzimologia
11.
Am J Pathol ; 184(10): 2757-67, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25131752

RESUMO

Macrophage accumulation is one of the hallmarks of progressive kidney disease. Tissue-type plasminogen activator (tPA) is known to promote macrophage infiltration and renal inflammation during chronic kidney injury. However, the underlying mechanism remains largely unknown. We examined the role of tPA in macrophage motility in vivo by tracking fluorescence-labeled bone marrow-derived macrophages, and found that tPA-deficient mice had markedly fewer infiltrating fluorescence-labeled macrophages than the wild-type (WT) mice. Experiments in bone marrow chimeric mice further demonstrated that myeloid cells are the main source of endogenous tPA that promotes macrophage migration. In vitro studies showed that tPA promoted macrophage motility through its CD11b-mediated protease-independent function; and focal adhesion kinase (FAK), Rac-1, and NF-κB were indispensable to tPA-induced macrophage migration as either infection of FAK dominant-negative adenovirus or treatment with a Rac-1-specific inhibitor or NF-κB inhibitor abolished the effect of tPA. Moreover, ectopic FAK mimicked tPA and induced macrophage motility. tPA also activated migratory signaling in vivo. The accumulation of phospho-FAK-positive CD11b macrophages in the obstructed kidneys from WT mice was clearly attenuated in tPA knockout mice, which also displayed lower Rac-1 activity than their WT counterparts. Therefore, our results indicate that myeloid-derived tPA promotes macrophage migration through a novel signaling cascade involving FAK, Rac-1, and NF-κB.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Macrófagos/fisiologia , NF-kappa B/metabolismo , Neuropeptídeos/metabolismo , Transdução de Sinais , Ativador de Plasminogênio Tecidual/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Quimera , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo
12.
Am J Kidney Dis ; 63(4): 567-72, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24200462

RESUMO

BACKGROUND: The study of novel urinary biomarkers of acute kidney injury has expanded exponentially. Effective interpretation of data and meaningful comparisons between studies require awareness of factors that can adversely affect measurement. We examined how variations in short-term storage and processing might affect the measurement of urine biomarkers. STUDY DESIGN: Cross-sectional prospective. SETTING & PARTICIPANTS: Hospitalized patients from 2 sites: Yale New Haven Hospital (n=50) and University of California, San Francisco Medical Center (n=36). PREDICTORS: We tested the impact of 3 urine processing conditions on these biomarkers: (1) centrifugation and storage at 4°C for 48 hours before freezing at -80°C, (2) centrifugation and storage at 25°C for 48 hours before freezing at -80°C, and (3) uncentrifuged samples immediately frozen at -80°C. OUTCOMES: Urine concentrations of 5 biomarkers: neutrophil gelatinase-associated lipocalin (NGAL), interleukin 18 (IL-18), kidney injury molecule 1 (KIM-1), liver-type fatty acid-binding protein (L-FABP), and cystatin C. MEASUREMENTS: We measured urine biomarkers by established enzyme-linked immunosorbent assay methods. Biomarker values were log-transformed, and agreement with a reference standard of immediate centrifugation and storage at -80°C was compared using concordance correlation coefficients (CCCs). RESULTS: Neither storing samples at 4°C for 48 hours nor centrifugation had a significant effect on measured levels, with CCCs higher than 0.9 for all biomarkers tested. For samples stored at 25°C for 48 hours, excellent CCC values (>0.9) also were noted between the test sample and the reference standard for NGAL, cystatin C, L-FABP and KIM-1. However, the CCC for IL-18 between samples stored at 25°C for 48 hours and the reference standard was 0.81 (95% CI, 0.66-0.96). LIMITATIONS: No comparisons to fresh, unfrozen samples; no evaluation of the effect of protease inhibitors. CONCLUSIONS: All candidate markers tested using the specified assays showed high stability with both short-term storage at 4°C and without centrifugation prior to freezing. For optimal fidelity, urine for IL-18 measurement should not be stored at 25°C before long-term storage or analysis.


Assuntos
Injúria Renal Aguda/urina , Biomarcadores/urina , Idoso , Idoso de 80 Anos ou mais , Centrifugação , Estudos Transversais , Ensaio de Imunoadsorção Enzimática , Feminino , Congelamento , Humanos , Masculino , Estudos Prospectivos , Coleta de Urina/métodos , Coleta de Urina/normas
13.
Am J Physiol Renal Physiol ; 304(5): F515-21, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23283990

RESUMO

Diabetes is a risk factor for the development of acute kidney injury (AKI) in humans and rodents. However, the mechanistic basis for this observation is unknown. The present studies evaluated the role of inflammation and TNF-α in ischemic AKI in a model of type 2 diabetes mellitus (DM). Diabetic (db/db) and nondiabetic (db/+) littermates were subjected to 20 min of bilateral renal ischemia. The nondiabetic mice developed only mild and transient renal dysfunction. In contrast, the equivalent ischemic insult provoked severe and sustained renal dysfunction in the db/db mice. The expression of TNF-α and Toll-like receptor 4 (TLR4) mRNA was measured in the kidneys of diabetic mice before and after renal ischemia; db/db mice exhibited greater increases in TNF-α and TLR4 mRNA expression following ischemia than did db/+. In addition, urinary excretion of TNF-α after ischemia was higher in db/db mice than in db/+ mice. To determine the possible role of TNF-α in mediating the enhanced susceptibility of diabetic mice to ischemic injury, db/db mice were injected with either a neutralizing anti-mouse TNF-α antibody or nonimmune globulin and then subjected to 20 min of bilateral renal ischemia. Treatment of the db/db mice with the TNF-α antibody provided significant protection against the ischemic injury. These data support the view that diabetes increases the susceptibility to ischemia-induced renal dysfunction. This increased susceptibility derives from a heightened inflammatory response involving TNF-α and perhaps TLR4 signaling.


Assuntos
Injúria Renal Aguda/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Isquemia/metabolismo , Rim/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Injúria Renal Aguda/complicações , Injúria Renal Aguda/fisiopatologia , Animais , Anticorpos Neutralizantes , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/fisiopatologia , Suscetibilidade a Doenças , Inflamação/complicações , Inflamação/metabolismo , Inflamação/fisiopatologia , Isquemia/complicações , Isquemia/fisiopatologia , Rim/irrigação sanguínea , Rim/fisiopatologia , Masculino , Camundongos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/genética
14.
BMC Nephrol ; 11: 22, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20799966

RESUMO

BACKGROUND: The incidence of acute kidney injury (AKI) has been increasing over time and is associated with a high risk of short-term death. Previous studies on hospital-acquired AKI have important methodological limitations, especially their retrospective study designs and limited ability to control for potential confounding factors. METHODS: The Assessment, Serial Evaluation, and Subsequent Sequelae of Acute Kidney Injury (ASSESS-AKI) Study was established to examine how a hospitalized episode of AKI independently affects the risk of chronic kidney disease development and progression, cardiovascular events, death, and other important patient-centered outcomes. This prospective study will enroll a cohort of 1100 adult participants with a broad range of AKI and matched hospitalized participants without AKI at three Clinical Research Centers, as well as 100 children undergoing cardiac surgery at three Clinical Research Centers. Participants will be followed for up to four years, and will undergo serial evaluation during the index hospitalization, at three months post-hospitalization, and at annual clinic visits, with telephone interviews occurring during the intervening six-month intervals. Biospecimens will be collected at each visit, along with information on lifestyle behaviors, quality of life and functional status, cognitive function, receipt of therapies, interim renal and cardiovascular events, electrocardiography and urinalysis. CONCLUSIONS: ASSESS-AKI will characterize the short-term and long-term natural history of AKI, evaluate the incremental utility of novel blood and urine biomarkers to refine the diagnosis and prognosis of AKI, and identify a subset of high-risk patients who could be targeted for future clinical trials to improve outcomes after AKI.


Assuntos
Injúria Renal Aguda/fisiopatologia , Projetos de Pesquisa , Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/mortalidade , Injúria Renal Aguda/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Biomarcadores/urina , Criança , Pré-Escolar , Estudos de Coortes , Coleta de Dados/métodos , Interpretação Estatística de Dados , Progressão da Doença , Humanos , Lactente , Falência Renal Crônica , Pessoa de Meia-Idade , Seleção de Pacientes , Prognóstico , Estudos Prospectivos , Tempo , Adulto Jovem
15.
Am J Pathol ; 177(4): 1687-96, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20724593

RESUMO

In renal fibrosis, interstitial fibroblasts have an increased proliferative phenotype, and the numbers of interstitial fibroblasts closely correlate with the extent of kidney damage. The mechanisms underlying proliferation and resulting expansion of the interstitium remain largely unknown. Here we define the intracellular signaling events by which tissue plasminogen activator (tPA) promotes renal interstitial fibroblast proliferation. tPA promoted the proliferation of renal interstitial fibroblasts independent of its protease activity. The mitogenic effect of tPA required Tyr(4507) phosphorylation of the cytoplasmic tail of its receptor LDL receptor-related protein 1. tPA triggered sequential proliferative signaling events involving Erk1/2, p90RSK, GSK3ß phosphorylation, and cyclin D1 induction. Blockade of Erk1/2 activation or knockdown of p90RSK suppressed tPA-induced GSK3ß phosphorylation, cyclin D1 expression, and fibroblast proliferation. In contrast, expression of constitutively active Mek1 mimicked tPA in inducing GSK3ß phosphorylation and cyclin D1 expression. Ectopic overexpression of an uninhibitable GSK3ß mutant eliminated tPA-induced cyclin D1 expression. In the murine obstruction model, tPA deficiency reduced renal GSK3ß phosphorylation and induction of PCNA and FSP-1. These findings show that tPA induces Tyr(4507) phosphorylation of LDL receptor-related protein 1, which in turn leads to the downstream phosphorylation of Erk1/2, p90RSK, and GSK3ß, followed by the induction of cyclin D1 in murine interstitial fibroblasts. This study implicates tPA as a mitogen that promotes interstitial fibroblast proliferation, leading to expansion of these cells.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Mitose , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais , Ativador de Plasminogênio Tecidual/farmacologia , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Ciclina D1/metabolismo , Embrião de Mamíferos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Imunofluorescência , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Técnicas Imunoenzimáticas , Imunoprecipitação , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Fosforilação , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tirosina/metabolismo
16.
J Am Soc Nephrol ; 21(1): 53-63, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19875815

RESUMO

Inflammation contributes to the pathogenesis of acute kidney injury. Dendritic cells (DCs) are immune sentinels with the ability to induce immunity or tolerance, but whether they mediate acute kidney injury is unknown. Here, we studied the distribution of DCs within the kidney and the role of DCs in cisplatin-induced acute kidney injury using a mouse model in which DCs express both green fluorescence protein and the diphtheria toxin receptor. DCs were present throughout the tubulointerstitium but not in glomeruli. We used diphtheria toxin to deplete DCs to study their functional significance in cisplatin nephrotoxicity. Mice depleted of DCs before or coincident with cisplatin treatment but not at later stages experienced more severe renal dysfunction, tubular injury, neutrophil infiltration and greater mortality than nondepleted mice. We used bone marrow chimeric mice to confirm that the depletion of CD11c-expressing hematopoietic cells was responsible for the enhanced renal injury. Finally, mixed bone marrow chimeras demonstrated that the worsening of cisplatin nephrotoxicity in DC-depleted mice was not a result of the dying or dead DCs themselves. After cisplatin treatment, expression of MHC class II decreased and expression of inducible co-stimulator ligand increased on renal DCs. These data demonstrate that resident DCs reduce cisplatin nephrotoxicity and its associated inflammation.


Assuntos
Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Células Dendríticas/patologia , Células Dendríticas/fisiologia , Injúria Renal Aguda/induzido quimicamente , Animais , Apoptose/fisiologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Antígeno CD11c/metabolismo , Cisplatino , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Néfrons/patologia , Néfrons/fisiopatologia
17.
Am J Physiol Renal Physiol ; 296(4): F723-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19211685

RESUMO

The cellular hallmark of kidney repair is a rapid proliferation of renal tubular epithelial cells ultimately leading to the restoration of nephron structure and function. Netrin-1 was discovered as a neural guidance cue and found to be expressed outside the nervous system, including in kidney. Previous work showed that netrin-1 is upregulated in response to ischemic injury and ameliorates ischemic injury. The objectives of this study were to determine the role of netrin-1 in renal tubular epithelial cell proliferation and migration in vitro. Real-time RT-PCR analysis showed that netrin-1 and its receptors UNC5B and neogenin are highly expressed in cultured mouse renal epithelial cells (TKPTS), whereas the expression of the Deleted in Colon Cancer (DCC), UNC5A, UNC5C, and UNC5D receptors is negligible or undetectable. Netrin-1 protein was induced in the edges of mechanical wounds in vitro. Netrin-1 increased TKPTS cell proliferation in a dose-dependent manner. The netrin-1-induced increase in TKPTS cell proliferation was completely prevented by small interfering RNA (siRNA) inhibition of UNC5B receptor but not UNC5C receptor expression. Netrin-1 also increased TKPTS cell migration in vitro, and this was also mediated through the UNC5B receptor. Netrin-1 increased the phosphorylation of Akt and ERK. Inhibition of phosphatidylinositol 3-kinase and MEK1/2 completely inhibited netrin-1-induced cell proliferation but not migration. These results indicate that netrin-1 increases renal tubular epithelial cell proliferation and migration through the UNC5B receptor. Moreover, the increase in cell proliferation, but not migration, was mediated via activation of Akt and ERK pathways.


Assuntos
Movimento Celular , Proliferação de Células , Células Epiteliais/metabolismo , Túbulos Renais Proximais/metabolismo , Fatores de Crescimento Neural/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/enzimologia , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/metabolismo , Camundongos , Fatores de Crescimento Neural/genética , Receptores de Netrina , Netrina-1 , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores de Superfície Celular/genética , Proteínas Supressoras de Tumor/genética , Cicatrização
18.
Am J Physiol Renal Physiol ; 296(1): F135-44, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18971209

RESUMO

Meprin metalloproteases, composed of alpha and/or beta subunits, consist of membrane-bound and secreted forms that are abundantly expressed in proximal tubules of the kidney as well as secreted into the urinary tract. Previous studies indicated that meprin metalloproteases play a role in pathological conditions such as ischemic acute renal failure and urinary tract infection. The aim of this work was to examine the role of meprins in endotoxemic acute renal failure using meprin alpha knockout (alphaKO), meprin beta knockout (betaKO), and wild-type (WT) mice. Differences among the responses of the genotypes were observed as early as 1 h after challenge with 2.5 mg/kg ip Escherichia coli LPS, establishing roles for meprins in the endotoxemic response. Meprin alphaKO mice displayed lower blood urea nitrogen levels and decreased nitric oxide levels, indicative of a decreased systemic response to LPS compared with WT and meprin betaKO mice. Serum cytokine profiles showed lower levels of IL-1beta and TNF-alpha in the meprin alphaKO mice within 3 h after LPS challenge and confirmed a role for meprins in the early phases of the host response. Meprin alphaKO mice were also hyporesponsive to LPS administered to the bladder, exhibiting significantly less bladder edema, leukocyte infiltration, and bladder permeability than WT mice. These data indicate that meprin A contributes to the renal and urogenital pathogenesis of endotoxicity.


Assuntos
Injúria Renal Aguda/metabolismo , Inflamação/metabolismo , Túbulos Renais Proximais/metabolismo , Lipopolissacarídeos/efeitos adversos , Metaloendopeptidases/metabolismo , Bexiga Urinária/metabolismo , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Animais , Nitrogênio da Ureia Sanguínea , Modelos Animais de Doenças , Inflamação/etiologia , Inflamação/patologia , Interleucina-1beta/sangue , Túbulos Renais Proximais/patologia , Leucócitos/patologia , Masculino , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Fator de Necrose Tumoral alfa/sangue , Bexiga Urinária/patologia
19.
Anal Chem ; 80(17): 6594-601, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18681455

RESUMO

Early detection of disease markers can provide higher diagnostic power and improve disease prognosis. We demonstrate the use of zinc oxide nanorod (ZnO NR) arrays in a straightforward, reliable, and ultrasensitive detection of the cytokines interleukin-18 and tumor necrosis factor-alpha. Specifically, we exploit the fluorescence-enhancing properties of ZnO NR platforms in cytokine assays involving both a pure buffer and urine. The detection sensitivity achieved using this ZnO NR method is in the subfemtogram per milliliter level, which is 3-4 orders of magnitude more sensitive than conventional assay detection limits. This unparalleled detection sensitivity is achieved without the need for indirect enzyme reactions or specialized instrumentation. We highlight various advantages of using ZnO NR arrays in the ultrasensitive profiling of cytokine levels. Key advantages include robustness of NR arrays, simple and direct assay schemes, high-throughput and multiplexing capabilities, and the ability to correlate directly measured signals to cytokine levels. In conjunction with the extremely high sensitivity demonstrated in this work, our ZnO NR array-based approach may be highly beneficial in early detection of many cytokine-implicated diseases.


Assuntos
Citocinas/análise , Citocinas/urina , Análise em Microsséries/métodos , Nanotubos/química , Óxido de Zinco/química , Animais , Bovinos , Humanos , Interleucina-18/análise , Interleucina-18/urina , Sensibilidade e Especificidade , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/urina
20.
Am J Physiol Renal Physiol ; 294(4): F731-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18234954

RESUMO

Acute kidney injury is an important complication in hospitalized patients often diagnosed late and associated with high mortality and morbidity. Although biomarkers for nephrotoxicity are available, they often lack sensitivity and specificity for detecting tubular injury. Netrin-1 is a laminin-like molecule highly expressed in many organs including kidney. To determine the value of netrin-1 as a biomarker of renal injury, we analyzed its urinary excretion following ischemia-reperfusion-, cisplatin-, folic acid-, and endotoxin-induced renal injury in mice. Urinary netrin-1 levels increased markedly within 3 h of ischemia-reperfusion (40 +/- 14-fold, P < 0.01 vs. baseline), reached a peak level at 6 h, and decreased thereafter, returning to near baseline by 72 h. Serum creatinine significantly increased only after 24 h of reperfusion. Similarly, in cisplatin-, folic acid-, and lipopolysaccharide-treated mice, urine netrin-1 excretion increased as early as 1 h and reached a peak level at 6 h after injection. However, serum creatinine was raised significantly after 6, 24, and 72 h after folic acid, lipopolysaccharide, and cisplatin administration, respectively. NGAL excretion in folic acid- and lipopolysaccharide-treated mice urine samples could only be detected by 24 h after drug administration. Furthermore, urinary netrin-1 excretion increased dramatically in 13 acute renal failure patients, whereas none was detected in 6 healthy volunteer urine samples. Immunohistochemical localization showed that netrin-1 is highly expressed in tubular epithelial cells in transplanted human kidney. We conclude that urinary netrin-1 is a promising early biomarker of renal injury.


Assuntos
Injúria Renal Aguda/diagnóstico , Nefropatias/diagnóstico , Rim/lesões , Fatores de Crescimento Neural/urina , Proteínas Supressoras de Tumor/urina , Injúria Renal Aguda/induzido quimicamente , Proteínas de Fase Aguda , Animais , Biomarcadores/análise , Cisplatino/uso terapêutico , Creatinina/sangue , Humanos , Lipocalina-2 , Lipocalinas/sangue , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Netrina-1 , Proteínas Proto-Oncogênicas/sangue , Circulação Renal , Traumatismo por Reperfusão/induzido quimicamente , Traumatismo por Reperfusão/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA