Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cereb Blood Flow Metab ; 43(2_suppl): 37-48, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37434361

RESUMEN

Nicotinamide adenine dinucleotide (NAD+) is a redox cofactor critical for oxidative phosphorylation. Nicotinamide (NAM) and nicotinamide riboside (NR) are NAD+ precursors widely used as nutritional supplements to augment oxidative phosphorylation. Indeed, NAD+ precursors have been reported to improve outcomes in ischemic stroke when administered as a rescue therapy after stroke onset. However, we have also reported that enhanced reliance on oxidative phosphorylation before ischemia onset might worsen outcomes. To address the paradox, we examined how NAD+ precursors modulate the outcome of middle cerebral artery occlusion in mice, when administered either 20 minutes after reperfusion or daily for three days before ischemia onset. A single post-ischemic dose of NAM or NR indeed improved tissue and neurologic outcomes examined at 72 hours. In contrast, pre-ischemic treatment for three days enlarged the infarcts and worsened neurological deficits. As a possible explanation for the diametric outcomes, a single dose of NAM or NR augmented tissue AMPK, PGC1α, SIRT1, and ATP in both naïve and ischemic brains, while the multiple-dose paradigm failed to do so. Our data suggest that NAD+ precursor supplements may sensitize the brain to subsequent ischemic events, despite their neuroprotective effect when administered after ischemia onset.


Asunto(s)
NAD , Accidente Cerebrovascular , Ratones , Animales , NAD/metabolismo , Suplementos Dietéticos , Encéfalo/metabolismo , Accidente Cerebrovascular/metabolismo , Isquemia
2.
World J Clin Cases ; 11(17): 3980-3992, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37388789

RESUMEN

Kidney disease in patients with liver disease is serious and increases mortality. Up to 50% of patients hospitalized experience an episode of acute kidney injury. In general, men with liver disease are thought to be at increased risk of kidney disease. However, this association should be considered with caution because most studies use creatinine-based inclusion criteria, which is negatively biased against women. In this review, we synthesize data on sex differences in kidney disease in patients with chronic liver disease in the clinical setting and discuss potential physiologic underpinnings.

4.
Nat Rev Nephrol ; 16(2): 99-111, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31673160

RESUMEN

The mammalian kidney relies on abundant mitochondria in the renal tubule to generate sufficient ATP to provide the energy required for constant reclamation of solutes from crude blood filtrate. The highly metabolically active cells of the renal tubule also pair their energetic needs to the regulation of diverse cellular processes, including energy generation, antioxidant responses, autophagy and mitochondrial quality control. Nicotinamide adenine dinucleotide (NAD+) is essential not only for the harvesting of energy from substrates but also for an array of regulatory reactions that determine cellular health. In acute kidney injury (AKI), substantial decreases in the levels of NAD+ impair energy generation and, ultimately, the core kidney function of selective solute transport. Conversely, augmentation of NAD+ may protect the kidney tubule against diverse acute stressors. For example, NAD+ augmentation can ameliorate experimental AKI triggered by ischaemia-reperfusion, toxic injury and systemic inflammation. NAD+-dependent maintenance of renal tubular metabolic health may also attenuate long-term profibrotic responses that could lead to chronic kidney disease. Further understanding of the genetic, environmental and nutritional factors that influence NAD+ biosynthesis and renal resilience may lead to novel approaches for the prevention and treatment of kidney disease.


Asunto(s)
Lesión Renal Aguda/metabolismo , Túbulos Renales/metabolismo , NAD/metabolismo , Insuficiencia Renal Crónica/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Humanos , NADP/metabolismo , Oxidación-Reducción , Poli(ADP-Ribosa) Polimerasas/metabolismo , Daño por Reperfusión/metabolismo , Sirtuinas/metabolismo
5.
JCI Insight ; 52019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30870143

RESUMEN

Because injured mitochondria can accelerate cell death through the elaboration of oxidative free radicals and other mediators, it is striking that proliferator gamma coactivator 1-alpha (PGC1α), a stimulator of increased mitochondrial abundance, protects stressed renal cells instead of potentiating injury. Here we report that PGC1α's induction of lysosomes via transcription factor EB (TFEB) may be pivotal for kidney protection. CRISPR and stable gene transfer showed that PGC1α knockout tubular cells were sensitized to the genotoxic stressor cisplatin whereas transgenic cells were protected. The biosensor mtKeima unexpectedly revealed that cisplatin blunts mitophagy both in cells and mice. PGC1α not only counteracted this effect but also raised basal mitophagy, as did the downstream mediator nicotinamide adenine dinucleotide (NAD+). PGC1α did not consistently affect known autophagy pathways modulated by cisplatin. Instead RNA sequencing identified coordinated regulation of lysosomal biogenesis via TFEB. This effector pathway was sufficiently important that inhibition of TFEB or lysosomes unveiled a striking harmful effect of excess PGC1α in cells and conditional mice. These results uncover an unexpected effect of cisplatin on mitophagy and PGC1α's exquisite reliance on lysosomes for kidney protection. Finally, the data illuminate TFEB as a novel target for renal tubular stress resistance.


Asunto(s)
Lesión Renal Aguda/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Cisplatino/toxicidad , Túbulos Renales/metabolismo , Lisosomas/metabolismo , Mitocondrias/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Sistemas CRISPR-Cas , Técnicas de Transferencia de Gen , Túbulos Renales/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitofagia/genética , NAD/metabolismo , Estrés Oxidativo/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Análisis de Secuencia de ARN
6.
Nat Med ; 24(9): 1351-1359, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30127395

RESUMEN

Nicotinamide adenine dinucleotide (NAD+) extends longevity in experimental organisms, raising interest in its impact on human health. De novo NAD+ biosynthesis from tryptophan is evolutionarily conserved yet considered supplanted among higher species by biosynthesis from nicotinamide (NAM). Here we show that a bottleneck enzyme in de novo biosynthesis, quinolinate phosphoribosyltransferase (QPRT), defends renal NAD+ and mediates resistance to acute kidney injury (AKI). Following murine AKI, renal NAD+ fell, quinolinate rose, and QPRT declined. QPRT+/- mice exhibited higher quinolinate, lower NAD+, and higher AKI susceptibility. Metabolomics suggested an elevated urinary quinolinate/tryptophan ratio (uQ/T) as an indicator of reduced QPRT. Elevated uQ/T predicted AKI and other adverse outcomes in critically ill patients. A phase 1 placebo-controlled study of oral NAM demonstrated a dose-related increase in circulating NAD+ metabolites. NAM was well tolerated and was associated with less AKI. Therefore, impaired NAD+ biosynthesis may be a feature of high-risk hospitalizations for which NAD+ augmentation could be beneficial.


Asunto(s)
Lesión Renal Aguda/metabolismo , Vías Biosintéticas , NAD/biosíntesis , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/orina , Anciano , Animales , Procedimientos Quirúrgicos Cardíacos , Humanos , Isquemia/orina , Ratones , Persona de Mediana Edad , Niacinamida/administración & dosificación , Niacinamida/uso terapéutico , Pentosiltransferasa/metabolismo , Proyectos Piloto , Ácido Quinolínico/metabolismo , Ácido Quinolínico/orina , Resultado del Tratamiento , Triptófano/orina
7.
Kidney Int ; 90(3): 638-47, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27282937

RESUMEN

Immune checkpoint inhibitors (CPIs), monoclonal antibodies that target inhibitory receptors expressed on T cells, represent an emerging class of immunotherapy used in treating solid organ and hematologic malignancies. We describe the clinical and histologic features of 13 patients with CPI-induced acute kidney injury (AKI) who underwent kidney biopsy. Median time from initiation of a CPI to AKI was 91 (range, 21 to 245) days. Pyuria was present in 8 patients, and the median urine protein to creatinine ratio was 0.48 (range, 0.12 to 0.98) g/g. An extrarenal immune-related adverse event occurred prior to the onset of AKI in 7 patients. Median peak serum creatinine was 4.5 (interquartile range, 3.6-7.3) mg/dl with 4 patients requiring hemodialysis. The prevalent pathologic lesion was acute tubulointerstitial nephritis in 12 patients, with 3 having granulomatous features, and 1 thrombotic microangiopathy. Among the 12 patients with acute tubulointerstitial nephritis, 10 received treatment with glucocorticoids, resulting in complete or partial improvement in renal function in 2 and 7 patients, respectively. However, the 2 patients with acute tubulointerstitial nephritis not given glucocorticoids had no improvement in renal function. Thus, CPI-induced AKI is a new entity that presents with clinical and histologic features similar to other causes of drug-induced acute tubulointerstitial nephritis, though with a longer latency period. Glucocorticoids appear to be a potentially effective treatment strategy. Hence, AKI due to CPIs may be caused by a unique mechanism of action linked to reprogramming of the immune system, leading to loss of tolerance.


Asunto(s)
Lesión Renal Aguda/patología , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos/efectos adversos , Factores Inmunológicos/antagonistas & inhibidores , Inmunoterapia/efectos adversos , Neoplasias/tratamiento farmacológico , Nefritis Intersticial/patología , Microangiopatías Trombóticas/patología , Lesión Renal Aguda/sangre , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/terapia , Adulto , Anciano , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Biopsia , Creatinina/sangre , Femenino , Glucocorticoides/uso terapéutico , Humanos , Inmunoterapia/métodos , Riñón/irrigación sanguínea , Riñón/patología , Pruebas de Función Renal , Masculino , Persona de Mediana Edad , Neoplasias/inmunología , Nefritis Intersticial/sangre , Nefritis Intersticial/inducido químicamente , Nefritis Intersticial/terapia , Diálisis Renal , Microangiopatías Trombóticas/sangre , Microangiopatías Trombóticas/inducido químicamente , Microangiopatías Trombóticas/terapia
8.
Semin Nephrol ; 36(1): 8-16, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27085731

RESUMEN

Acute kidney injury (AKI) continues to be a significant contributor to morbidity, mortality, and health care expenditure. In the United States alone, it is estimated that more than $10 billion is spent on AKI every year. Currently, there are no available therapies to treat established AKI. The mitochondrion is positioned to be a critical player in AKI with its dual role as the primary source of energy for each cell and as a key regulator of cell death. This review aims to cover the current state of research on the role of mitochondria in AKI, while also proposing potential therapeutic targets and future therapies.


Asunto(s)
Lesión Renal Aguda/metabolismo , Mitocondrias/fisiología , Humanos , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...