Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 104(6): 1194-1205, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37652206

RESUMEN

Biomarkers of tubular function such as epidermal growth factor (EGF) may improve prognostication of participants at highest risk for chronic kidney disease (CKD) after hospitalization. To examine this, we measured urinary EGF (uEGF) from samples collected in the Assessment, Serial Evaluation, and Subsequent Sequelae of Acute Kidney Injury (ASSESS-AKI) Study, a multi-center, prospective, observational cohort of hospitalized participants with and without AKI. Cox proportional hazards regression was used to investigate the association of uEGF/Cr at hospitalization, three months post-discharge, and the change between these time points with major adverse kidney events (MAKE): CKD incidence, progression, or development of kidney failure. Clinical findings were paired with mechanistic studies comparing relative Egf expression in mouse models of kidney atrophy or repair after ischemia-reperfusion injury. MAKE was observed in 20% of 1,509 participants over 4.3 years of follow-up. Each 2-fold higher level of uEGF/Cr at three months was associated with decreased risk of MAKE (adjusted hazards ratio 0.46, 95% confidence interval: 0.39-0.55). Participants with the highest increase in uEGF/Cr from hospitalization to three-month follow-up had a lower risk of MAKE (adjusted hazards ratio 0.52; 95% confidence interval: 0.36-0.74) compared to those with the least change in uEGF/Cr. A model using uEGF/Cr at three months combined with clinical variables yielded moderate discrimination for MAKE (area under the curve 0.73; 95% confidence interval: 0.69-0.77) and strong discrimination for kidney failure at four years (area under the curve 0.96; 95% confidence interval: 0.92-1.00). Accelerated restoration of Egf expression in mice was seen in the model of adaptive repair after injury, compared to a model of progressive atrophy. Thus, urinary EGF/Cr may be a biomarker of distal tubular health, with higher concentrations and increased uEGF/Cr post-discharge independently associated with reduced risk of MAKE in hospitalized patients.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Humanos , Animales , Ratones , Factor de Crecimiento Epidérmico , Estudios Prospectivos , Cuidados Posteriores , Tasa de Filtración Glomerular , Alta del Paciente , Riñón , Insuficiencia Renal Crónica/diagnóstico , Biomarcadores , Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/epidemiología , Atrofia
2.
J Am Soc Nephrol ; 33(6): 1077-1086, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35577558

RESUMEN

BACKGROUND: After kidney injury, macrophages transition from initial proinflammatory activation to a proreparative phenotype characterized by expression of arginase-1 (Arg1), mannose receptor 1 (Mrc1), and macrophage scavenger receptor 1 (Msr1). The mechanism by which these alternatively activated macrophages promote repair is unknown. METHODS: We characterized the macrophage and renal responses after ischemia-reperfusion injury with contralateral nephrectomy in LysM-Cre;Arg1fl/fl mice and littermate controls and used in vitro coculture of macrophages and tubular cells to determine how macrophage-expressed arginase-1 promotes kidney repair. RESULTS: After ischemia-reperfusion injury with contralateral nephrectomy, Arg1-expressing macrophages were almost exclusively located in the outer stripe of the medulla adjacent to injured S3 tubule segments containing luminal debris or casts. Macrophage Arg1 expression was reduced by more than 90% in injured LysM-Cre;Arg1fl/fl mice, resulting in decreased mouse survival, decreased renal tubular cell proliferation and decreased renal repair compared with littermate controls. In vitro studies demonstrate that tubular cells exposed apically to dead cell debris secrete high levels of GM-CSF and induce reparative macrophage activation, with those macrophages in turn secreting Arg1-dependent factor(s) that directly stimulate tubular cell proliferation. CONCLUSIONS: GM-CSF-induced, proreparative macrophages express arginase-1, which is required for the S3 tubular cell proliferative response that promotes renal repair after ischemia-reperfusion injury.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Daño por Reperfusión , Animales , Arginasa/genética , Arginasa/metabolismo , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Regeneración , Daño por Reperfusión/metabolismo
3.
Am J Physiol Renal Physiol ; 322(3): F322-F334, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35100823

RESUMEN

Renal tubular casts originating from detached epithelial cells after ischemia-reperfusion injury (IRI) can obstruct tubules and negatively impact glomerular filtration rate. Using multiphoton imaging of 400-µm-thick kidney sections, the distribution of casts and morphometric measurement of tubules was performed along the entire nephron for the first time. Tubular nuclei are shed before cell detachment, and visually occlusive casts (grade 3) appeared at 12 h after IRI at the S3/thin descending limb (tDL) junction. Grade 3 casts peaked at 24 h after injury [present in 99% of S3, 78% of tDL, 76% of thin ascending limb (tAL), 60% of medullary thick ascending limb (mTAL), and 10% of connecting tubule segments]. Cast formation in the S3 correlated with selective loss of cell numbers from this tubule segment. By day 3, most mTALs and connecting tubules were cast free, whereas 72% of S3 tubules and 58% of tDLs still contained grade 3 casts. Although bulk phagocytosis of cast material by surviving tubular cells was not observed, mass spectrometry identified large numbers of tryptic peptides in the outer medulla, and trypsin levels were significantly increased in the kidney and urine 24 h after IRI. Administration of either antipain or camostat to inhibit trypsin extended cast burden to the S2, led to sustained accumulation of S3 casts after IRI, but did not affect cast burden in the mTAL or renal function. Our data provide detailed and dynamic mapping of tubular cast formation and resolution after IRI that can inform future interventions to accelerate cast clearance and renal recovery.NEW & NOTEWORTHY This detailed characterization of the dynamic distribution of dead cell debris in ischemically injured kidney tubules reveals which cells in the kidney are most severely injured, when and where tubular casts form, and when (and to a lesser extent, how) they are cleared.


Asunto(s)
Nefronas , Daño por Reperfusión , Tasa de Filtración Glomerular , Humanos , Riñón , Túbulos Renales
4.
Nephrol Dial Transplant ; 37(11): 2214-2222, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34865148

RESUMEN

BACKGROUND: Patients with acute interstitial nephritis (AIN) can present without typical clinical features, leading to a delay in diagnosis and treatment. We therefore developed and validated a diagnostic model to identify patients at risk of AIN using variables from the electronic health record. METHODS: In patients who underwent a kidney biopsy at Yale University between 2013 and 2018, we tested the association of >150 variables with AIN, including demographics, comorbidities, vital signs and laboratory tests (training set 70%). We used least absolute shrinkage and selection operator methodology to select prebiopsy features associated with AIN. We performed area under the receiver operating characteristics curve (AUC) analysis with internal (held-out test set 30%) and external validation (Biopsy Biobank Cohort of Indiana). We tested the change in model performance after the addition of urine biomarkers in the Yale AIN study. RESULTS: We included 393 patients (AIN 22%) in the training set, 158 patients (AIN 27%) in the test set, 1118 patients (AIN 11%) in the validation set and 265 patients (AIN 11%) in the Yale AIN study. Variables in the selected model included serum creatinine {adjusted odds ratio [aOR] 2.31 [95% confidence interval (CI) 1.42-3.76]}, blood urea nitrogen:creatinine ratio [aOR 0.40 (95% CI 0.20-0.78)] and urine dipstick specific gravity [aOR 0.95 (95% CI 0.91-0.99)] and protein [aOR 0.39 (95% CI 0.23-0.68)]. This model showed an AUC of 0.73 (95% CI 0.64-0.81) in the test set, which was similar to the AUC in the external validation cohort [0.74 (95% CI 0.69-0.79)]. The AUC improved to 0.84 (95% CI 0.76-0.91) upon the addition of urine interleukin-9 and tumor necrosis factor-α. CONCLUSIONS: We developed and validated a statistical model that showed a modest AUC for AIN diagnosis, which improved upon the addition of urine biomarkers. Future studies could evaluate this model and biomarkers to identify unrecognized cases of AIN.


Asunto(s)
Interleucina-9 , Nefritis Intersticial , Humanos , Creatinina , Interleucina-9/uso terapéutico , Registros Electrónicos de Salud , Factor de Necrosis Tumoral alfa , Nefritis Intersticial/diagnóstico , Nefritis Intersticial/epidemiología , Nefritis Intersticial/tratamiento farmacológico , Biopsia , Biomarcadores/análisis
5.
Am J Kidney Dis ; 77(4): 490-499.e1, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33422598

RESUMEN

RATIONALE & OBJECTIVE: Although coronavirus disease 2019 (COVID-19) has been associated with acute kidney injury (AKI), it is unclear whether this association is independent of traditional risk factors such as hypotension, nephrotoxin exposure, and inflammation. We tested the independent association of COVID-19 with AKI. STUDY DESIGN: Multicenter, observational, cohort study. SETTING & PARTICIPANTS: Patients admitted to 1 of 6 hospitals within the Yale New Haven Health System between March 10, 2020, and August 31, 2020, with results for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing via polymerase chain reaction of a nasopharyngeal sample. EXPOSURE: Positive test for SARS-CoV-2. OUTCOME: AKI by KDIGO (Kidney Disease: Improving Global Outcomes) criteria. ANALYTICAL APPROACH: Evaluated the association of COVID-19 with AKI after controlling for time-invariant factors at admission (eg, demographic characteristics, comorbidities) and time-varying factors updated continuously during hospitalization (eg, vital signs, medications, laboratory results, respiratory failure) using time-updated Cox proportional hazard models. RESULTS: Of the 22,122 patients hospitalized, 2,600 tested positive and 19,522 tested negative for SARS-CoV-2. Compared with patients who tested negative, patients with COVID-19 had more AKI (30.6% vs 18.2%; absolute risk difference, 12.5% [95% CI, 10.6%-14.3%]) and dialysis-requiring AKI (8.5% vs 3.6%) and lower rates of recovery from AKI (58% vs 69.8%). Compared with patients without COVID-19, patients with COVID-19 had higher inflammatory marker levels (C-reactive protein, ferritin) and greater use of vasopressors and diuretic agents. Compared with patients without COVID-19, patients with COVID-19 had a higher rate of AKI in univariable analysis (hazard ratio, 1.84 [95% CI, 1.73-1.95]). In a fully adjusted model controlling for demographic variables, comorbidities, vital signs, medications, and laboratory results, COVID-19 remained associated with a high rate of AKI (adjusted hazard ratio, 1.40 [95% CI, 1.29-1.53]). LIMITATIONS: Possibility of residual confounding. CONCLUSIONS: COVID-19 is associated with high rates of AKI not fully explained by adjustment for known risk factors. This suggests the presence of mechanisms of AKI not accounted for in this analysis, which may include a direct effect of COVID-19 on the kidney or other unmeasured mediators. Future studies should evaluate the possible unique pathways by which COVID-19 may cause AKI.


Asunto(s)
Lesión Renal Aguda/epidemiología , COVID-19/epidemiología , Lesión Renal Aguda/sangre , Lesión Renal Aguda/terapia , Anciano , Proteína C-Reactiva/metabolismo , COVID-19/metabolismo , COVID-19/terapia , Estudios de Cohortes , Creatinina/sangre , Diuréticos/uso terapéutico , Femenino , Mortalidad Hospitalaria , Humanos , Unidades de Cuidados Intensivos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Diálisis Renal , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/epidemiología , Respiración Artificial , Factores de Riesgo , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Estados Unidos/epidemiología , Vasoconstrictores/uso terapéutico
6.
Nephrol Dial Transplant ; 36(10): 1851-1858, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-33125471

RESUMEN

BACKGROUND: We previously demonstrated that urine interleukin (IL)-9 and tumor necrosis factor (TNF)-α can distinguish acute interstitial nephritis (AIN) from other causes of acute kidney injury. Here we evaluated the role of these biomarkers to prognosticate kidney function in patients with AIN. METHODS: In a cohort of participants with biopsy-proven, adjudicated AIN, we tested the association of histological features and urine biomarkers (IL-9 and TNF-α) with estimated glomerular filtration rate measured 6 months after diagnosis (6 m-eGFR) controlling for eGFR before AIN and albuminuria. We also evaluated subgroups in whom corticosteroid use was associated with 6 m-eGFR. RESULTS: In the 51 (93%) of the 55 participants with complete data, median (interquartile range) eGFR before and 6 m after AIN were 41 (27-69) and 28 (13-47) mL/min/1.73 m2, respectively. Patients with higher severity of interstitial fibrosis had lower 6 m-eGFR, whereas those with higher tubulointerstitial infiltrate had higher 6 m-eGFR. IL-9 levels were associated with lower 6 m-eGFR only in the subset of patients who did not receive corticosteroids [6m-eGFR per doubling of IL-9, -6.0 (-9.4 to -2.6) mL/min/1.73 m2]. Corticosteroid use was associated with higher 6 m-eGFR [20.9 (0.2, 41.6) mL/min/1.73 m2] only in those with urine IL-9 above the median (>0.66 ng/g) but not in others. CONCLUSIONS: Urine IL-9 was associated with lower 6 m-eGFR only in participants not treated with corticosteroids. Corticosteroid use was associated with higher 6 m-eGFR in those with high urine IL-9. These findings provide a framework for IL-9-guided clinical trials to test efficacy of immunosuppressive therapy in patients with AIN.


Asunto(s)
Interleucina-9/orina , Nefritis Intersticial , Factor de Necrosis Tumoral alfa , Tasa de Filtración Glomerular , Humanos , Nefritis Intersticial/diagnóstico , Nefritis Intersticial/tratamiento farmacológico , Pronóstico , Factor de Necrosis Tumoral alfa/orina
7.
Clin Nephrol ; 96(2): 112-119, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34142944

RESUMEN

Retroperitoneal fibrosis and chronic periaortitis describe overlapping groups of rare diseases characterized by inflammation and fibrosis involving the aorta. The presentation is often non-specific, and while obstructive nephropathy is a common complication, these entities are an uncommon cause of renal failure necessitating dialysis. A 57-year-old man presented multiple times with acute kidney injury, even requiring hemodialysis, with repeated abrupt resolution. Renal ultrasound repeatedly did not reveal acute hydronephrosis. Renal biopsy on his first admission showed acute tubular injury attributed to hypovolemia. Computed tomography finally revealed a retroperitoneal soft tissue mass encasing the infrarenal abdominal aorta and partially encasing the bilateral ureters. Bilateral nephrostomy tubes were placed, steroids were initiated, and the patient experienced rapid and remarkable improvement in renal function. Chronic periaortitis should be considered in older patients with acute kidney injury, even in the absence of ultrasonographic evidence of obstruction. Additional studies are needed to describe the test characteristics of renal sonography for periaortitis, the long-term sequelae of acute kidney injury secondary to periaortitis, and the optimal management to preserve long-term renal function.


Asunto(s)
Lesión Renal Aguda , Fibrosis Retroperitoneal , Humanos , Riñón/patología , Masculino , Persona de Mediana Edad
8.
Annu Rev Physiol ; 79: 449-469, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28192060

RESUMEN

Acute kidney injury (AKI) is a growing global health concern, yet no treatment is currently available to prevent it or to promote kidney repair after injury. Animal models demonstrate that the macrophage is a major contributor to the inflammatory response to AKI. Emerging data from human biopsies also corroborate the presence of macrophages in AKI and their persistence in progressive chronic kidney disease. Macrophages are phagocytic innate immune cells that are important mediators of tissue homeostasis and host defense. In response to tissue injury, macrophages become activated based on specific signals from the damaged microenvironment. The activation and functional state of the macrophage depends on the stage of tissue injury and repair, reflecting a dynamic and diverse spectrum of macrophage phenotypes. In this review, we highlight our current understanding of the mechanisms by which macrophages contribute to injury and repair after AKI.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Riñón/fisiopatología , Macrófagos/fisiología , Animales , Microambiente Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata/fisiología , Inflamación/patología
9.
J Am Soc Nephrol ; 30(10): 1825-1840, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31315923

RESUMEN

BACKGROUND: After bilateral kidney ischemia/reperfusion injury (IRI), monocytes infiltrate the kidney and differentiate into proinflammatory macrophages in response to the initial kidney damage, and then transition to a form that promotes kidney repair. In the setting of unilateral IRI (U-IRI), however, we have previously shown that macrophages persist beyond the time of repair and may promote fibrosis. METHODS: Macrophage homing/survival signals were determined at 14 days after injury in mice subjected to U-IRI and in vitro using coculture of macrophages and tubular cells. Mice genetically engineered to lack Ccr2 and wild-type mice were treated ±CCR2 antagonist RS102895 and subjected to U-IRI to quantify macrophage accumulation, kidney fibrosis, and inflammation 14 and 30 days after the injury. RESULTS: Failure to resolve tubular injury after U-IRI results in sustained expression of granulocyte-macrophage colony-stimulating factor by renal tubular cells, which directly stimulates expression of monocyte chemoattractant protein-1 (Mcp-1) by macrophages. Analysis of CD45+ immune cells isolated from wild-type kidneys 14 days after U-IRI reveals high-level expression of the MCP-1 receptor Ccr2. In mice lacking Ccr2 and wild-type mice treated with RS102895, the numbers of macrophages, dendritic cells, and T cell decreased following U-IRI, as did the expression of profibrotic growth factors and proimflammatory cytokines. This results in a reduction in extracellular matrix and kidney injury markers. CONCLUSIONS: GM-CSF-induced MCP-1/CCR2 signaling plays an important role in the cross-talk between injured tubular cells and infiltrating immune cells and myofibroblasts, and promotes sustained inflammation and tubular injury with progressive interstitial fibrosis in the late stages of U-IRI.


Asunto(s)
Quimiocina CCL2/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Inflamación/etiología , Riñón/irrigación sanguínea , Riñón/patología , Receptores CCR2/fisiología , Daño por Reperfusión/complicaciones , Animales , Células Cultivadas , Fibrosis/etiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Túbulos Renales/citología , Túbulos Renales/metabolismo , Macrófagos , Ratones
10.
J Am Soc Nephrol ; 29(10): 2471-2481, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30209078

RESUMEN

BACKGROUND: In patients with autosomal dominant polycystic kidney disease (ADPKD), most of whom have a mutation in PKD1 or PKD2, abnormally large numbers of macrophages accumulate around kidney cysts and promote their growth. Research by us and others has suggested that monocyte chemoattractant protein-1 (Mcp1) may be a signal for macrophage-mediated cyst growth. METHODS: To define the role of Mcp1 and macrophages in promoting cyst growth, we used mice with inducible knockout of Pkd1 alone (single knockout) or knockout of both Pkd1 and Mcp1 (double knockout) in the murine renal tubule. Levels of Mcp1 RNA expression were measured in single-knockout mice and controls. RESULTS: In single-knockout mice, upregulation of Mcp1 precedes macrophage infiltration. Macrophages accumulating around nascent cysts (0-2 weeks after induction) are initially proinflammatory and induce tubular cell injury with morphologic flattening, oxidative DNA damage, and proliferation-independent cystic dilation. At 2-6 weeks after induction, macrophages switch to an alternative activation phenotype and promote further cyst growth because of an additional three-fold increase in tubular cell proliferative rates. In double-knockout mice, there is a marked reduction in Mcp1 expression and macrophage numbers, resulting in less initial tubular cell injury, slower cyst growth, and improved renal function. Treatment of single-knockout mice with an inhibitor to the Mcp1 receptor Ccr2 partially reproduced the morphologic and functional improvement seen with Mcp1 knockout. CONCLUSIONS: Mcp1 is upregulated after knockout of Pkd1 and promotes macrophage accumulation and cyst growth via both proliferation-independent and proliferation-dependent mechanisms in this orthologous mouse model of ADPKD.


Asunto(s)
Quimiocina CCL2/genética , Quimiocina CCL2/fisiología , Macrófagos/fisiología , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Animales , Quimiocina CCL2/deficiencia , Daño del ADN , Modelos Animales de Enfermedad , Humanos , Túbulos Renales/patología , Túbulos Renales/fisiopatología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética , Activación de Macrófagos/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Riñón Poliquístico Autosómico Dominante/fisiopatología , Pirrolidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptores CCR2/antagonistas & inhibidores , Canales Catiónicos TRPP/deficiencia , Canales Catiónicos TRPP/genética , Regulación hacia Arriba
11.
J Am Soc Nephrol ; 28(11): 3218-3226, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28679671

RESUMEN

The normal response to kidney injury includes a robust inflammatory infiltrate of PMNs and macrophages. We previously showed that the small secreted protein breast regression protein-39 (BRP-39), also known as chitinase 3-like 1 (CHI3L1) and encoded by the Chi3l1 gene, is expressed at high levels by macrophages during the early stages of kidney repair and promotes tubular cell survival via IL-13 receptor α2 (IL13Rα2)-mediated signaling. Here, we investigated the role of BRP-39 in profibrotic responses after AKI. In wild-type mice, failure to resolve tubular injury after unilateral ischemia-reperfusion injury (U-IRI) led to sustained low-level Chi3l1 mRNA expression by renal cells and promoted macrophage persistence and severe interstitial fibrosis. Analysis of macrophages isolated from wild-type kidneys 14 days after U-IRI revealed high-level expression of the profibrotic BRP-39 receptor Ptgdr2/Crth2 and expression of the profibrotic markers Lgals3, Pdgfb, Egf, and Tgfb In comparison, injured kidneys from mice lacking BRP-39 had significantly fewer macrophages, reduced expression of profibrotic growth factors, and decreased accumulation of extracellular matrix. BRP-39 depletion did not affect myofibroblast accumulation but did attenuate myofibroblast expression of Col1a1, Col3a1, and Fn1 Together, these results identify BRP-39 as an important activator of macrophage-myofibroblast crosstalk and profibrotic signaling in the setting of maladaptive kidney repair.


Asunto(s)
Lesión Renal Aguda/etiología , Proteína 1 Similar a Quitinasa-3/fisiología , Riñón/patología , Miofibroblastos/fisiología , Animales , Fibrosis/etiología , Masculino , Ratones
13.
J Am Soc Nephrol ; 28(2): 661-670, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27451287

RESUMEN

Deceased donor kidneys with AKI are often discarded for fear of poor transplant outcomes. Donor biomarkers that predict post-transplant renal recovery could improve organ selection and reduce discard. We tested whether higher levels of donor urinary YKL-40, a repair phase protein, associate with improved recipient outcomes in a prospective cohort study involving deceased kidney donors from five organ procurement organizations. We measured urinary YKL-40 concentration in 1301 donors (111 had AKI, defined as doubling of serum creatinine) and ascertained outcomes in the corresponding 2435 recipients, 756 of whom experienced delayed graft function (DGF). Donors with AKI had higher urinary YKL-40 concentration (P<0.001) and acute tubular necrosis on procurement biopsies (P=0.05). In fully adjusted analyses, elevated donor urinary YKL-40 concentration associated with reduced risk of DGF in both recipients of AKI donor kidneys (adjusted relative risk, 0.51 [95% confidence interval (95% CI), 0.32 to 0.80] for highest versus lowest YKL-40 tertile) and recipients of non-AKI donor kidneys (adjusted relative risk, 0.79 [95% CI, 0.65 to 0.97]). Furthermore, in the event of DGF, elevated donor urinary YKL-40 concentration associated with higher 6-month eGFR (6.75 [95% CI, 1.49 to 12.02] ml/min per 1.73 m2) and lower risk of graft failure (adjusted hazard ratio, 0.50 [95% CI, 0.27 to 0.94]). These findings suggest that YKL-40 is produced in response to tubular injury and is independently associated with recovery from AKI and DGF. If ultimately validated as a prognostic biomarker, urinary YKL-40 should be considered in determining the suitability of donor kidneys for transplant.


Asunto(s)
Lesión Renal Aguda/orina , Proteína 1 Similar a Quitinasa-3/orina , Funcionamiento Retardado del Injerto/epidemiología , Trasplante de Riñón , Adulto , Cadáver , Femenino , Humanos , Masculino , Estudios Prospectivos , Recuperación de la Función , Donantes de Tejidos , Obtención de Tejidos y Órganos
14.
J Am Soc Nephrol ; 26(6): 1334-45, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25388222

RESUMEN

After kidney ischemia/reperfusion (I/R) injury, monocytes home to the kidney and differentiate into activated macrophages. Whereas proinflammatory macrophages contribute to the initial kidney damage, an alternatively activated phenotype can promote normal renal repair. The microenvironment of the kidney during the repair phase mediates the transition of macrophage activation from a proinflammatory to a reparative phenotype. In this study, we show that macrophages isolated from murine kidneys during the tubular repair phase after I/R exhibit an alternative activation gene profile that differs from the canonical alternative activation induced by IL-4-stimulated STAT6 signaling. This unique activation profile can be reproduced in vitro by stimulation of bone marrow-derived macrophages with conditioned media from serum-starved mouse proximal tubule cells. Secreted tubular factors were found to activate macrophage STAT3 and STAT5 but not STAT6, leading to induction of the unique alternative activation pattern. Using STAT3-deficient bone marrow-derived macrophages and pharmacologic inhibition of STAT5, we found that tubular cell-mediated macrophage alternative activation is regulated by STAT5 activation. Both in vitro and after renal I/R, tubular cells expressed GM-CSF, a known STAT5 activator, and this pathway was required for in vitro alternative activation of macrophages by tubular cells. Furthermore, administration of a neutralizing antibody against GM-CSF after renal I/R attenuated kidney macrophage alternative activation and suppressed tubular proliferation. Taken together, these data show that tubular cells can instruct macrophage activation by secreting GM-CSF, leading to a unique macrophage reparative phenotype that supports tubular proliferation after sterile ischemic injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Activación de Macrófagos/genética , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/fisiopatología , Análisis de Varianza , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunohistoquímica , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis Multivariante , Fenotipo , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Daño por Reperfusión/fisiopatología , Transducción de Señal , Regulación hacia Arriba
15.
J Biol Chem ; 289(20): 14341-50, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24692544

RESUMEN

While Wnt and Hgf signaling pathways are known to regulate epithelial cell responses during injury and repair, whether they exhibit functional cross-talk is not well defined. Canonical Wnt signaling is initiated by the phosphorylation of the Lrp5/6 co-receptors. In the current study we demonstrate that Hgf stimulates Met and Gsk3-dependent and Wnt-independent phosphorylation of Lrp5/6 at three separate activation motifs in subconfluent, de-differentiated renal epithelial cells. Hgf treatment stimulates the selective association of active Gsk3 with Lrp5/6. In contrast, Akt-phosphorylated inactive Gsk3 is excluded from this association. Hgf stimulates ß-catenin stabilization and nuclear accumulation and protects against epithelial cell apoptosis in an Lrp5/6-dependent fashion. In vivo, the increase in Lrp5/6 phosphorylation and ß-catenin stabilization in the first 6-24 h after renal ischemic injury was significantly reduced in mice lacking Met receptor in the renal proximal tubule. Our results thus identify Hgf as an important transactivator of canonical Wnt signaling that is mediated by Met-stimulated, Gsk3-dependent Lrp5/6 phosphorylation.


Asunto(s)
Factor de Crecimiento de Hepatocito/farmacología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Isquemia/metabolismo , Isquemia/patología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/patología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Fosforilación/efectos de los fármacos , beta Catenina/metabolismo
16.
Pediatr Nephrol ; 30(2): 199-209, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24442822

RESUMEN

Acute ischemic kidney injury is a common complication in hospitalized patients. No treatment is yet available for augmenting kidney repair or preventing progressive kidney fibrosis. Animal models of acute kidney injury demonstrate that activation of the innate immune system plays a major role in the systemic response to ischemia/reperfusion injury. Macrophage depletion studies suggest that macrophages, key participants in the innate immune response, augment the initial injury after reperfusion but also promote tubular repair and contribute to long-term kidney fibrosis after ischemic injury. The distinct functional outcomes seen following macrophage depletion at different time points after ischemia/reperfusion injury suggest heterogeneity in macrophage activation states. Identifying the pathways that regulate the transitions of macrophage activation is thus critical for understanding the mechanisms that govern both macrophage-mediated injury and repair in the postischemic kidney. This review examines our understanding of the complex and intricately controlled pathways that determine monocyte recruitment, macrophage activation, and macrophage effector functions after renal ischemia/reperfusion injury. Careful delineation of repair and resolution pathways could provide therapeutic targets for the development of effective treatments to offer patients with acute kidney injury.


Asunto(s)
Lesión Renal Aguda/inmunología , Macrófagos/inmunología , Daño por Reperfusión/inmunología , Animales , Humanos
17.
J Am Soc Nephrol ; 25(2): 329-37, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24136921

RESUMEN

Renal proximal tubule epithelial cells express high levels of the hepatocyte growth factor receptor Met, and both the receptor and ligand are upregulated after ischemic injury. Activation of the Met receptor after hepatocyte growth factor stimulation in vitro promotes activities involved in kidney repair, including cell survival, migration, and proliferation. However, characterizing the in vivo role of these signaling events in proximal tubule responses to kidney injury has been difficult because global Met knockout results in embryonic lethality due to placental and liver abnormalities. Here, we used γGT-Cre to knockout Met receptor expression selectively in the proximal tubules of mice (γGT-Cre;Met(fl/fl)). The kidneys of these mice developed normally, but exhibited increased initial tubular injury, tubular cell apoptosis, and serum creatinine after ischemia/reperfusion compared with γGT-Cre;Met(+/+) kidneys. These changes in γGT-Cre;Met(fl/fl) mice correlated with a selective reduction in PI3K/Akt activation in response to injury and subsequent decreases in inhibitory phosphorylation of the proapoptotic factor Bad and activating phosphorylation of the ribosomal regulatory protein p70-S6 kinase. Moreover, tubular cell proliferation after ischemia/reperfusion was delayed in γGT-Cre;Met(fl/fl) mice. In conclusion, this study identifies Met-dependent phosphoinositide 3-kinase activation in proximal tubules as a critical determinant of initial tubular cell survival and reparative proliferation after ischemic injury.


Asunto(s)
Lesión Renal Aguda/enzimología , Riñón/irrigación sanguínea , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/fisiología , Daño por Reperfusión/enzimología , Transducción de Señal/fisiología , Animales , Apoptosis , Técnicas de Silenciamiento del Gen , Riñón/enzimología , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Proteína Letal Asociada a bcl/metabolismo
18.
Respir Res ; 15: 32, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24641672

RESUMEN

BACKGROUND: Previous work in our laboratory demonstrated that hyperoxia suppressed the expression of vascular endothelial growth factor (VEGF) by the embryonic lung, leading to increased epithelial cell apoptosis and failure of explant airway growth and branching that was rescued by the addition of Vegf165. The aims of this study were to determine protective pathways by which VEGF isoforms attenuate hyperoxic lung growth retardation and to identify the target cell for VEGF action. METHODS: Timed pregnant CD-1 or fetal liver kinase (FLK1)-eGFP lung explants cultured in 3% or 50% oxygen were treated ± Vegf121, VEGF164/Vegf165 or VEGF188 in the presence or absence of anti-rat neuropilin-1 (NRP1) antibody or GO6983 (protein kinase C (PKC) pan-inhibitor) and lung growth and branching quantified. Immunofluorescence studies were performed to determine apoptosis index and location of FLK1 phosphorylation and western blot studies of lung explants were performed to define the signaling pathways that mediate the protective effects of VEGF. RESULTS: Heparin-binding VEGF isoforms (VEGF164/Vegf165 and VEGF188) but not Vegf121 selectively reduced epithelial apoptosis and partially rescued lung bud branching and growth. These protective effects required NRP1-dependent FLK1 activation in endothelial cells. Analysis of downstream signaling pathways demonstrated that the VEGF-mediated anti-apoptotic effects were dependent on PKC activation. CONCLUSIONS: Vegf165 activates FLK1-NRP1 signaling in endothelial cells, leading to a PKC-dependent paracrine signal that in turn inhibits epithelial cell apoptosis.


Asunto(s)
Heparina/metabolismo , Pulmón/metabolismo , Neuropilina-1/fisiología , Proteína Quinasa C/fisiología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , Apoptosis/fisiología , Hipoxia de la Célula/fisiología , Femenino , Pulmón/crecimiento & desarrollo , Pulmón/patología , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Embarazo , Unión Proteica/fisiología , Isoformas de Proteínas/fisiología , Distribución Aleatoria , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
BMC Nephrol ; 15: 133, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25128003

RESUMEN

BACKGROUND: A translational study in renal transplantation suggested YKL-40, a chitinase 3-like-1 gene product, plays an important role in acute kidney injury (AKI) and repair, but data are lacking about this protein in urine from native human kidneys. METHODS: This is an ancillary study to a single-center, prospective observational cohort of patients with clinically-defined AKI according to AKI Network serum creatinine criteria. We determined the association of YKL -40 ≥ 5 ng/ml, alone or combined with neutrophil gelatinase-associated lipocalin (NGAL), in urine collected on the first day of AKI with a clinically important composite outcome (progression to higher AKI stage and/or in-hospital death). RESULTS: YKL-40 was detectable in all 249 patients, but urinary concentrations were considerably lower than in previously measured deceased-donor kidney transplant recipients. Seventy-two patients (29%) progressed or died in-hospital, and YKL-40 ≥ 5 ng/ml had an adjusted odds ratio (95% confidence interval) for the outcome of 3.4 (1.5-7.7). The addition of YKL-40 to a clinical model for predicting the outcome resulted in a continuous net reclassification improvement of 29% (P = 0.04). In patients at high risk for the outcome based on NGAL concentrations in the upper quartile, YKL-40 further partitioned the cohort into moderate-risk and very high-risk groups. CONCLUSIONS: Urine YKL-40 is associated with AKI progression and/or death in hospitalized patients and improves clinically determined risk reclassification. Combining YKL-40 with other AKI biomarkers like NGAL may further delineate progression risk, though additional studies are needed to determine whether YKL-40 has general applicability and to define its association with longer-term outcomes in AKI.


Asunto(s)
Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/orina , Adipoquinas/orina , Progresión de la Enfermedad , Hospitalización , Lectinas/orina , Lesión Renal Aguda/mortalidad , Anciano , Biomarcadores/orina , Proteína 1 Similar a Quitinasa-3 , Estudios de Cohortes , Femenino , Hospitalización/tendencias , Humanos , Masculino , Persona de Mediana Edad , Mortalidad/tendencias , Estudios Prospectivos
20.
BMC Nephrol ; 15: 105, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24996668

RESUMEN

BACKGROUND: Adjudication of patient outcomes is a common practice in medical research and clinical trials. However minimal data exists on the adjudication process in the setting of Acute Kidney Injury (AKI) as well as the ability to judge different etiologies (e.g. Acute Tubular Necrosis (ATN), Pre-renal Azotemia (PRA)). METHODS: We enrolled 475 consecutive patients undergoing cardiac surgery at four sites of the Translational Research Investigating Biomarker Endpoints in AKI (TRIBE-AKI) study. Three expert nephrologists performed independent chart review, utilizing clinical variables and retrospective case report forms with pre intra and post-operative data, and then adjudicated all cases of AKI (n = 67). AKI was defined as a > 50% increase in serum creatinine for baseline (RIFLE Risk). We examined the patterns of AKI diagnoses made by the adjudication panel as well as association of these diagnoses with pre and postoperative kidney injury biomarkers. RESULTS: There was poor agreement across the panel of reviewers with their adjudicated diagnoses being independent of each other (Fleiss' Kappa = 0.046). Based on the agreement of the two out of three reviewers, ATN was the adjudicated diagnosis in 41 cases (61%) while PRA occurred in 13 (19%). Neither serum creatinine or any other biomarker of AKI (urine or serum), was associated with an adjudicated diagnosis of ATN within the first 24 post-operative hours. CONCLUSION: The etiology of AKI after cardiac surgery is probably multi-factorial and pure forms of AKI etiologies, such as ATN and PRA may not exist. Biomarkers did not appear to correlate with the adjudicated etiology of AKI; however the lack of agreement among the adjudicators impacted these results. TRIAL REGISTRATION: Clinicaltrials.gov: NCT00774137.


Asunto(s)
Lesión Renal Aguda/sangre , Lesión Renal Aguda/etiología , Puente de Arteria Coronaria/efectos adversos , Complicaciones Posoperatorias/sangre , Complicaciones Posoperatorias/etiología , Investigación Biomédica Traslacional/métodos , Lesión Renal Aguda/diagnóstico , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/diagnóstico , Estudios Prospectivos , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA