Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
JCI Insight ; 9(11)2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38855868

RESUMEN

Lactate elevation is a well-characterized biomarker of mitochondrial dysfunction, but its role in diabetic kidney disease (DKD) is not well defined. Urine lactate was measured in patients with type 2 diabetes (T2D) in 3 cohorts (HUNT3, SMART2D, CRIC). Urine and plasma lactate were measured during euglycemic and hyperglycemic clamps in participants with type 1 diabetes (T1D). Patients in the HUNT3 cohort with DKD had elevated urine lactate levels compared with age- and sex-matched controls. In patients in the SMART2D and CRIC cohorts, the third tertile of urine lactate/creatinine was associated with more rapid estimated glomerular filtration rate decline, relative to first tertile. Patients with T1D demonstrated a strong association between glucose and lactate in both plasma and urine. Glucose-stimulated lactate likely derives in part from proximal tubular cells, since lactate production was attenuated with sodium-glucose cotransporter-2 (SGLT2) inhibition in kidney sections and in SGLT2-deficient mice. Several glycolytic genes were elevated in human diabetic proximal tubules. Lactate levels above 2.5 mM potently inhibited mitochondrial oxidative phosphorylation in human proximal tubule (HK2) cells. We conclude that increased lactate production under diabetic conditions can contribute to mitochondrial dysfunction and become a feed-forward component to DKD pathogenesis.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Glucólisis , Ácido Láctico , Humanos , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Animales , Ratones , Ácido Láctico/metabolismo , Ácido Láctico/sangre , Femenino , Masculino , Persona de Mediana Edad , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Mitocondrias/metabolismo , Adulto , Tasa de Filtración Glomerular , Anciano , Túbulos Renales Proximales/metabolismo , Glucosa/metabolismo , Fosforilación Oxidativa , Biomarcadores/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Transportador 2 de Sodio-Glucosa/genética , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología
2.
Cell Rep ; 42(3): 112155, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36857182

RESUMEN

The most abundant cellular divalent cations, Mg2+ (mM) and Ca2+ (nM-µM), antagonistically regulate divergent metabolic pathways with several orders of magnitude affinity preference, but the physiological significance of this competition remains elusive. In mice consuming a Western diet, genetic ablation of the mitochondrial Mg2+ channel Mrs2 prevents weight gain, enhances mitochondrial activity, decreases fat accumulation in the liver, and causes prominent browning of white adipose. Mrs2 deficiency restrains citrate efflux from the mitochondria, making it unavailable to support de novo lipogenesis. As citrate is an endogenous Mg2+ chelator, this may represent an adaptive response to a perceived deficit of the cation. Transcriptional profiling of liver and white adipose reveals higher expression of genes involved in glycolysis, ß-oxidation, thermogenesis, and HIF-1α-targets, in Mrs2-/- mice that are further enhanced under Western-diet-associated metabolic stress. Thus, lowering mMg2+ promotes metabolism and dampens diet-induced obesity and metabolic syndrome.


Asunto(s)
Tejido Adiposo Pardo , Metabolismo Energético , Animales , Ratones , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteínas de Transporte de Catión , Dieta , Dieta Alta en Grasa , Metabolismo Energético/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales , Obesidad/metabolismo , Termogénesis/genética
3.
Kidney Int Rep ; 7(7): 1502-1513, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35812275

RESUMEN

Introduction: Biomarkers of acute kidney injury (AKI) are often indexed to urine creatinine (UCr) or urine osmolarity (UOsm) to control for urine concentration. We evaluated how these approaches affect the biomarker-outcome association in patients with AKI. Methods: The Assessment, Serial Evaluation, and Subsequent Sequelae in Acute Kidney Injury Study was a cohort of hospitalized patients with and without AKI between 2009 and 2015. Using Cox proportional hazards regression, we assessed the associations and predictions (C-statistics) of urine biomarkers with a composite outcome of incident chronic kidney disease (CKD) and CKD progression. We used 4 approaches to account for urine concentration: indexing and adjusting for UCr and UOsm. Results: Among 1538 participants, 769 (50%) had AKI and 300 (19.5%) developed composite CKD outcome at median follow-up of 4.7 years. UCr and UOsm during hospitalization were inversely associated with the composite CKD outcome. The associations and predictions with CKD were significantly strengthened after indexing or adjusting for UCr or UOsm for urine kidney injury molecule-1 (KIM-1), interleukin-18 (IL-18), and monocyte chemoattractant protein-1 (MCP-1) in patients with AKI. There was no significant improvement with indexing or adjusting UCr or UOsm for albumin, neutrophil gelatinase-associated lipocalin (NGAL), and chitinase 3-like 1 (YKL-40). Uromodulin's (UMOD) inverse association with the outcome was significantly blunted after indexing but not adjusting for UCr or UOsm. Conclusion: UCr and UOsm during hospitalization are inversely associated with development and progression of CKD. Indexing or adjusting for UCr or UOsm strengthened associations and improved predictions for CKD for only some biomarkers. Incorporating urinary concentration should be individualized for each biomarker in research and clinical applications.

4.
J Am Soc Nephrol ; 33(3): 613-627, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35017169

RESUMEN

BACKGROUND: The mechanisms underlying long-term sequelae after AKI remain unclear. Vessel instability, an early response to endothelial injury, may reflect a shared mechanism and early trigger for CKD and heart failure. METHODS: To investigate whether plasma angiopoietins, markers of vessel homeostasis, are associated with CKD progression and heart failure admissions after hospitalization in patients with and without AKI, we conducted a prospective cohort study to analyze the balance between angiopoietin-1 (Angpt-1), which maintains vessel stability, and angiopoietin-2 (Angpt-2), which increases vessel destabilization. Three months after discharge, we evaluated the associations between angiopoietins and development of the primary outcomes of CKD progression and heart failure and the secondary outcome of all-cause mortality 3 months after discharge or later. RESULTS: Median age for the 1503 participants was 65.8 years; 746 (50%) had AKI. Compared with the lowest quartile, the highest quartile of the Angpt-1:Angpt-2 ratio was associated with 72% lower risk of CKD progression (adjusted hazard ratio [aHR], 0.28; 95% confidence interval [CI], 0.15 to 0.51), 94% lower risk of heart failure (aHR, 0.06; 95% CI, 0.02 to 0.15), and 82% lower risk of mortality (aHR, 0.18; 95% CI, 0.09 to 0.35) for those with AKI. Among those without AKI, the highest quartile of Angpt-1:Angpt-2 ratio was associated with 71% lower risk of heart failure (aHR, 0.29; 95% CI, 0.12 to 0.69) and 68% less mortality (aHR, 0.32; 95% CI, 0.15 to 0.68). There were no associations with CKD progression. CONCLUSIONS: A higher Angpt-1:Angpt-2 ratio was strongly associated with less CKD progression, heart failure, and mortality in the setting of AKI.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Cardíaca , Insuficiencia Renal Crónica , Lesión Renal Aguda/complicaciones , Anciano , Angiopoyetinas , Femenino , Insuficiencia Cardíaca/complicaciones , Humanos , Masculino , Pronóstico , Estudios Prospectivos , Insuficiencia Renal Crónica/complicaciones , Factores de Riesgo
5.
iScience ; 25(1): 103722, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35005527

RESUMEN

SARS-CoV-2 is a newly identified coronavirus that causes the respiratory disease called coronavirus disease 2019 (COVID-19). With an urgent need for therapeutics, we lack a full understanding of the molecular basis of SARS-CoV-2-induced cellular damage and disease progression. Here, we conducted transcriptomic analysis of human PBMCs, identified significant changes in mitochondrial, ion channel, and protein quality-control gene products. SARS-CoV-2 proteins selectively target cellular organelle compartments, including the endoplasmic reticulum and mitochondria. M-protein, NSP6, ORF3A, ORF9C, and ORF10 bind to mitochondrial PTP complex components cyclophilin D, SPG-7, ANT, ATP synthase, and a previously undescribed CCDC58 (coiled-coil domain containing protein 58). Knockdown of CCDC58 or mPTP blocker cyclosporin A pretreatment enhances mitochondrial Ca2+ retention capacity and bioenergetics. SARS-CoV-2 infection exacerbates cardiomyocyte autophagy and promotes cell death that was suppressed by cyclosporin A treatment. Our findings reveal that SARS-CoV-2 viral proteins suppress cardiomyocyte mitochondrial function that disrupts cardiomyocyte Ca2+ cycling and cell viability.

6.
Nat Rev Nephrol ; 17(5): 319-334, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547417

RESUMEN

Diabetic kidney disease is the leading cause of kidney failure worldwide; in the USA, it accounts for over 50% of individuals entering dialysis or transplant programmes. Unlike other complications of diabetes, the prevalence of diabetic kidney disease has failed to decline over the past 30 years. Hyperglycaemia is the primary aetiological factor responsible for the development of diabetic kidney disease. Once hyperglycaemia becomes established, multiple pathophysiological disturbances, including hypertension, altered tubuloglomerular feedback, renal hypoxia, lipotoxicity, podocyte injury, inflammation, mitochondrial dysfunction, impaired autophagy and increased activity of the sodium-hydrogen exchanger, contribute to progressive glomerular sclerosis and the decline in glomerular filtration rate. The quantitative contribution of each of these abnormalities to the progression of diabetic kidney disease, as well as their role in type 1 and type 2 diabetes mellitus, remains to be determined. Sodium-glucose co-transporter 2 (SGLT2) inhibitors have a beneficial impact on many of these pathophysiological abnormalities; however, as several pathophysiological disturbances contribute to the onset and progression of diabetic kidney disease, multiple agents used in combination will likely be required to slow the progression of disease effectively.


Asunto(s)
Nefropatías Diabéticas/etiología , Hipoglucemiantes/farmacología , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Autofagia/efectos de los fármacos , Nefropatías Diabéticas/prevención & control , Endotelio Vascular/efectos de los fármacos , Tasa de Filtración Glomerular/efectos de los fármacos , Humanos , Hiperglucemia/complicaciones , Hipertensión/complicaciones , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Obesidad/complicaciones , Podocitos/patología , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
7.
Cell ; 183(2): 474-489.e17, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33035451

RESUMEN

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.


Asunto(s)
Retículo Endoplásmico/metabolismo , Ácido Láctico/metabolismo , Magnesio/metabolismo , Animales , Células COS , Calcio/metabolismo , Señalización del Calcio/fisiología , Chlorocebus aethiops , Retículo Endoplásmico/fisiología , Femenino , Células HeLa , Células Hep G2 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo
8.
PLoS One ; 15(9): e0238816, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32898157

RESUMEN

Interleukin-10 (IL-10), a cytokine with anti-inflammatory effects, is produced by renal parenchymal cells and bone marrow derived cells. Both endogenous and exogenous IL-10 are protective in cisplatin-induced acute kidney injury. However, the source of endogenous IL-10 in cisplatin-induced nephrotoxicity is not clear. Bone marrow chimera experiments in IL10-KO mice indicated that bone marrow derived cells were the primary source of IL-10 in cisplatin nephrotoxicity. Cell specific deletion of IL-10 in T regulatory cells and dendritic cells was accomplished using Foxp3 and CD11c driven cre recombination in IL10flox/flox mice, respectively. Upon treatment with cisplatin, both the IL10flox/flox and the Foxp3YFP-Cre x IL10flox/flox mice developed similar degrees of kidney injury. However, mice with the dendritic cell deletion of IL-10 showed more severe structural and functional changes in the kidney compared to the IL10flox/flox mice. These results indicate that IL-10 from dendritic cells but not from T regulatory cells offers significant endogenous protection against cisplatin induced nephrotoxicity.


Asunto(s)
Cisplatino/efectos adversos , Células Dendríticas/metabolismo , Interleucina-10/metabolismo , Riñón/citología , Riñón/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Riñón/inmunología , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL
9.
JCI Insight ; 5(19)2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-32956070

RESUMEN

Fibrosis is the final common pathway in the pathophysiology of most forms of chronic kidney disease (CKD). As treatment of renal fibrosis still remains largely supportive, a refined understanding of the cellular and molecular mechanisms of kidney fibrosis and the development of novel compounds are urgently needed. Whether arginases play a role in the development of fibrosis in CKD is unclear. We hypothesized that endothelial arginase-2 (Arg2) promotes the development of kidney fibrosis induced by unilateral ureteral obstruction (UUO). Arg2 expression and arginase activity significantly increased following renal fibrosis. Pharmacologic blockade or genetic deficiency of Arg2 conferred kidney protection following renal fibrosis, as reflected by a reduction in kidney interstitial fibrosis and fibrotic markers. Selective deletion of Arg2 in endothelial cells (Tie2Cre/Arg2fl/fl) reduced the level of fibrosis after UUO. In contrast, selective deletion of Arg2 specifically in proximal tubular cells (Ggt1Cre/Arg2fl/fl) failed to reduce renal fibrosis after UUO. Furthermore, arginase inhibition restored kidney nitric oxide (NO) levels, oxidative stress, and mitochondrial function following UUO. These findings indicate that endothelial Arg2 plays a major role in renal fibrosis via its action on NO and mitochondrial function. Blocking Arg2 activity or expression could be a novel therapeutic approach for prevention of CKD.


Asunto(s)
Arginasa/antagonistas & inhibidores , Células Endoteliales/metabolismo , Fibrosis/prevención & control , Enfermedades Renales/prevención & control , Túbulos Renales Proximales/metabolismo , Obstrucción Ureteral/complicaciones , Animales , Arginasa/fisiología , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Estrés Oxidativo
10.
Sci Signal ; 13(628)2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32317369

RESUMEN

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.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Ácidos Grasos/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Ácido Pirúvico/metabolismo , Animales , Transporte Biológico Activo/genética , Canales de Calcio/genética , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Ratones Noqueados , Mitocondrias Hepáticas/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/genética
11.
JAMA Intern Med ; 180(3): 402-410, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31985750

RESUMEN

Importance: Among patients who had acute kidney injury (AKI) during hospitalization, there is a need to improve risk prediction such that those at highest risk for subsequent loss of kidney function are identified for appropriate follow-up. Objective: To evaluate the association of post-AKI proteinuria with increased risk of future loss of renal function. Design, Setting, and Participants: The Assessment, Serial Evaluation, and Subsequent Sequelae in Acute Kidney Injury (ASSESS-AKI) Study was a multicenter prospective cohort study including 4 clinical centers in North America included 1538 patients enrolled 3 months after hospital discharge between December 2009 and February 2015. Exposures: Urine albumin-to-creatinine ratio (ACR) quantified 3 months after hospital discharge. Main Outcomes and Measures: Kidney disease progression defined as halving of estimated glomerular filtration rate (eGFR) or end-stage renal disease. Results: Of the 1538 participants, 769 (50%) had AKI durring hospitalization. The baseline study visit took place at a mean (SD) 91 (23) days after discharge. The mean (SD) age was 65 (13) years; the median eGFR was 68 mL/min/1.73 m2; and the median urine ACR was 15 mg/g. Overall, 547 (37%) study participants were women and 195 (13%) were black. After a median follow-up of 4.7 years, 138 (9%) participants had kidney disease progression. Higher post-AKI urine ACR level was associated with increased risk of kidney disease progression (hazard ratio [HR], 1.53 for each doubling; 95% CI, 1.45-1.62), and urine ACR measurement was a strong discriminator for future kidney disease progression (C statistic, 0.82). The performance of urine ACR was stronger in patients who had had AKI than in those who had not (C statistic, 0.70). A comprehensive model of clinical risk factors (eGFR, blood pressure, and demographics) including ACR provided better discrimination for predicting kidney disease progression after hospital discharge among those who had had AKI (C statistic, 0.85) vs those who had not (C statistic, 0.76). In the entire matched cohort, after taking into account urine ACR, eGFR, demographics, and traditional chronic kidney risk factors determined 3 months after discharge, AKI (HR, 1.46; 95% CI, 0.51-4.13 for AKI vs non-AKI) or severity of AKI (HR, 1.54; 95% CI, 0.50-4.72 for AKI stage 1 vs non-AKI; HR, 0.56; 95% CI, 0.07-4.84 for AKI stage 2 vs non-AKI; HR, 2.24; 95% CI, 0.33-15.29 for AKI stage 3 vs non-AKI) was not independently associated with more rapid kidney disease progression. Conclusions and Relevance: Proteinuria level is a valuable risk-stratification tool in the post-AKI period. These results suggest there should be more widespread and routine quantification of proteinuria after hospitalized AKI.


Asunto(s)
Lesión Renal Aguda/complicaciones , Fallo Renal Crónico/etiología , Proteinuria/etiología , Lesión Renal Aguda/fisiopatología , Anciano , Presión Sanguínea/fisiología , Progresión de la Enfermedad , Femenino , Tasa de Filtración Glomerular/fisiología , Humanos , Fallo Renal Crónico/fisiopatología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Proteinuria/fisiopatología
12.
Trans Am Clin Climatol Assoc ; 130: 33-40, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31516162

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/inmunología , Células Dendríticas/inmunología , Inmunidad Innata/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Lesión Renal Aguda/inducido químicamente , Animales , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Modelos Animales de Enfermedad , Riñón/inmunología , Ratones
13.
J Am Assoc Lab Anim Sci ; 58(1): 21-29, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30538007

RESUMEN

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.


Asunto(s)
Anestesia General/veterinaria , Anestésicos/administración & dosificación , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Lipocalina 2/orina , Anestesia General/efectos adversos , Anestésicos/efectos adversos , Animales , Biomarcadores/orina , Regulación de la Expresión Génica/efectos de los fármacos , Riñón/efectos de los fármacos , Ciencia de los Animales de Laboratorio , Masculino , Ratones , Ratones Endogámicos C57BL , Estudios Prospectivos
14.
Kidney Int ; 93(2): 365-374, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29061334

RESUMEN

Ischemia/reperfusion is a common cause of acute kidney injury (AKI). However, mechanisms underlying the sudden loss in kidney function and tissue injury remain to be fully elucidated. Here, we investigated the role of peptidyl arginine deiminase-4 (PAD4), which converts arginine to citrulline and plays a role in epigenetic regulation and inflammation, in renal ischemia/reperfusion injury. PAD4 expression was highly induced in infiltrating leukocytes 24 hours following renal ischemia and reperfusion. This induction was accompanied by citrullination of histone H3 and formation of neutrophil extracellular traps in kidneys of wild-type mice. By contrast, PAD4-deficient mice did not form neutrophil extracellular traps, expressed lower levels of pro-inflammatory cytokines and were partially protected from renal ischemia/reperfusion-induced AKI. Furthermore, PAD4-deficient mice recovered kidney function 48 hours after ischemia/reperfusion, whereas kidney function in the wild-type mice progressively worsened. Administration of DNase I, which degrades neutrophil extracellular traps or the PAD-specific inhibitor YW3-56 before ischemia, partially prevented renal ischemia/reperfusion-induced AKI. Notably, transfer of neutrophils from wild-type, but not from PAD4-deficient mice, was sufficient to restore renal neutrophil extracellular trap formation and impair kidney function following renal ischemia/reperfusion. Thus, neutrophil PAD4 plays a pivotal role in renal ischemia/reperfusion-induced AKI.


Asunto(s)
Lesión Renal Aguda/enzimología , Trampas Extracelulares/enzimología , Hidrolasas/metabolismo , Riñón/enzimología , Neutrófilos/enzimología , Daño por Reperfusión/enzimología , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/prevención & control , Animales , Citrulinación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Histiocitos/metabolismo , Hidrolasas/antagonistas & inhibidores , Hidrolasas/deficiencia , Hidrolasas/genética , Mediadores de Inflamación/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Neutrófilos/trasplante , Arginina Deiminasa Proteína-Tipo 4 , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/prevención & control
15.
Kidney Int ; 92(1): 11-13, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28646989

RESUMEN

Inflammation is an important mediator of most forms of acute kidney injury (AKI). Although neutrophils are prominent components of the inflammatory cascade, the precise role of neutrophils in AKI and the mechanisms by which they contribute to AKI remain controversial. In this issue, Deng et al. identify an important cross talk between renal epithelial cells and neutrophils involving the production and action of leukotriene B4 in mediating cisplatin AKI. We discuss the possible explanations for the discrepant findings that have been reported for neutrophils in cisplatin AKI.


Asunto(s)
Cisplatino , Neutrófilos , Lesión Renal Aguda , Biomarcadores , Humanos , Inflamación
16.
Am J Physiol Renal Physiol ; 313(2): F522-F534, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28515179

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/enzimología , Arginasa/metabolismo , Daño por Reperfusión/enzimología , Lesión Renal Aguda/patología , Adenosina Trifosfato/metabolismo , Animales , Arginasa/antagonistas & inhibidores , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitritos/metabolismo , Estrés Oxidativo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Daño por Reperfusión/patología
17.
Kidney Int ; 91(5): 998-1000, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28407885

RESUMEN

Fructose consumption has been linked to hypertension in animal models and human studies, and endogenous fructose metabolism has been shown to promote acute and chronic kidney injury in mice. A recent study published in Nature Communications demonstrates a reduction in ischemic acute kidney injury with genetic knockout or inhibition of fructokinase, which catalyzes the first step in fructose metabolism. Although the role of this pathway in human kidney disease remains unclear, the recent description of several candidate fructokinase inhibitors may allow for clinical studies in the future.


Asunto(s)
Lesión Renal Aguda , Fructosa , Animales , Humanos , Riñón , Ratones
18.
Sensors (Basel) ; 17(2)2017 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-28241443

RESUMEN

Protein biomarkers, especially cytokines, play a pivotal role in the diagnosis and treatment of a wide spectrum of diseases. Therefore, a critical need for advanced cytokine sensors has been rapidly growing and will continue to expand to promote clinical testing, new biomarker development, and disease studies. In particular, sensors employing transduction principles of various optical modalities have emerged as the most common means of detection. In typical cytokine assays which are based on the binding affinities between the analytes of cytokines and their specific antibodies, optical schemes represent the most widely used mechanisms, with some serving as the gold standard against which all existing and new sensors are benchmarked. With recent advancements in nanoscience and nanotechnology, many of the recently emerging technologies for cytokine detection exploit various forms of nanomaterials for improved sensing capabilities. Nanomaterials have been demonstrated to exhibit exceptional optical properties unique to their reduced dimensionality. Novel sensing approaches based on the newly identified properties of nanomaterials have shown drastically improved performances in both the qualitative and quantitative analyses of cytokines. This article brings together the fundamentals in the literature that are central to different optical modalities developed for cytokine detection. Recent advancements in the applications of novel technologies are also discussed in terms of those that enable highly sensitive and multiplexed cytokine quantification spanning a wide dynamic range. For each highlighted optical technique, its current detection capabilities as well as associated challenges are discussed. Lastly, an outlook for nanomaterial-based cytokine sensors is provided from the perspective of optimizing the technologies for sensitivity and multiplexity as well as promoting widespread adaptations of the emerging optical techniques by lowering high thresholds currently present in the new approaches.


Asunto(s)
Técnicas Biosensibles , Citocinas , Humanos , Nanoestructuras , Nanotecnología
19.
Kidney Dis (Basel) ; 3(3): 120-126, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29344507

RESUMEN

BACKGROUND: Acute kidney injury (AKI) is a complex disease burdened by uncertainties of definition, management strategies, and prognosis. This study explores the relationship between demographic characteristics of nephrologists and their perceptions about the definition, management, and follow-up of AKI. METHODS: We developed a Web-based survey, the International Survey on Acute Kidney Injury (ISAKI), consisting of 29 items in 4 categories: (1) demographic and practice characteristics, (2) definition of AKI, (3) management of renal replacement therapy (RRT) in AKI, and (4) sequelae of AKI. A multivariable stepwise logistic regression model was used to examine relationships between the dependent variables and the demographic characteristics of the respondents. RESULTS: Responses from 743 nephrologists from 90 countries were analyzed. The majority (60%) of respondents reported using RIFLE and/or AKIN criteria regularly to define AKI, although US nephrologists were less likely to do so (OR: 0.58; 95% CI: 0.42-0.85). The most common initial RRT modality was intermittent hemodialysis (63.5%), followed by continuous RRT (23.8%). Faculty affiliation was associated with a higher likelihood of using a dialysis schedule of ≥4 times a week (OR: 1.75; 95% CI: 1.20-2.55). The respondents believed that a single episode of AKI increases the likelihood of development of chronic kidney disease (CKD) (55%), subsequent AKI (36%), and rapid progression of preexisting CKD (87%). US nephrologists were less likely to recommend follow-up after resolution of AKI (OR: 0.15; 95% CI: 0.07-0.33). CONCLUSIONS: Our findings highlight the need for a widely accepted consensus definition of AKI, a uniform approach to management, and improved follow-up after resolution of AKI episodes.

20.
Kidney Int ; 91(3): 671-682, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27914709

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Nefropatías Diabéticas/metabolismo , Podocitos/metabolismo , Receptores CCR2/metabolismo , Albuminuria/genética , Albuminuria/metabolismo , Albuminuria/prevención & control , Animales , Apoptosis , Nitrógeno de la Urea Sanguínea , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/prevención & control , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Monocitos/metabolismo , Fenotipo , Podocitos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR2/deficiencia , Receptores CCR2/genética , Transducción de Señal , Estreptozocina , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...