Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Physiol Genomics ; 56(1): 98-111, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37955135

RESUMEN

Salt sensitivity impacts a significant portion of the population and is an important contributor to the development of chronic kidney disease. One of the significant early predictors of salt-induced damage is albuminuria, which reflects the deterioration of the renal filtration barrier: the glomerulus. Despite significant research efforts, there is still a gap in knowledge regarding the molecular mechanisms and signaling networks contributing to instigating and/or perpetuating salt-induced glomerular injury. To address this gap, we used 8-wk-old male Dahl salt-sensitive rats fed a normal-salt diet (0.4% NaCl) or challenged with a high-salt diet (4% NaCl) for 3 wk. At the end of the protocol, a pure fraction of renal glomeruli obtained by differential sieving was used for next-generation RNA sequencing and comprehensive semi-automatic transcriptomic data analyses, which revealed 149 differentially expressed genes (107 and 42 genes were downregulated and upregulated, respectively). Furthermore, a combination of predictive gene correlation networks and computational bioinformatic analyses revealed pathways impacted by a high salt dietary challenge, including renal metabolism, mitochondrial function, apoptotic signaling and fibrosis, cell cycle, inflammatory and immune responses, circadian clock, cytoskeletal organization, G protein-coupled receptor signaling, and calcium transport. In conclusion, we report here novel transcriptomic interactions and corresponding predicted pathways affecting glomeruli under salt-induced stress.NEW & NOTEWORTHY Our study demonstrated novel pathways affecting glomeruli under stress induced by dietary salt. Predictive gene correlation networks and bioinformatic semi-automatic analysis revealed changes in the pathways relevant to mitochondrial function, inflammatory, apoptotic/fibrotic processes, and cell calcium transport.


Asunto(s)
Hipertensión , Cloruro de Sodio Dietético , Ratas , Animales , Masculino , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio/metabolismo , Hipertensión/genética , Ratas Endogámicas Dahl , Presión Sanguínea , Calcio/metabolismo , Transcriptoma/genética , Perfilación de la Expresión Génica , Riñón/metabolismo
2.
Am J Physiol Renal Physiol ; 325(2): F135-F149, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37262088

RESUMEN

Diabetic kidney disease (DKD) is one of the most devastating complications of diabetes mellitus, where currently there is no cure available. Several important mechanisms contribute to the pathogenesis of this complication, with oxidative stress being one of the key factors. The past decades have seen a large number of publications with various aspects of this topic; however, the specific details of redox regulation in DKD are still unclear. This is partly because redox biology is very complex, coupled with a complex and heterogeneous organ with numerous cell types. Furthermore, often times terms such as "oxidative stress" or reactive oxygen species are used as a general term to cover a wide and rich variety of reactive species and their differing reactions. However, no reactive species are the same, and not all of them are capable of biologically relevant reactions or "redox signaling." The goal of this review is to provide a biochemical background for an array of specific reactive oxygen species types with varying reactivity and specificity in the kidney as well as highlight some of the advances in redox biology that are paving the way to a better understanding of DKD development and risk.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Humanos , Nefropatías Diabéticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estrés Oxidativo/fisiología , Riñón/metabolismo , Oxidación-Reducción , Diabetes Mellitus/metabolismo
3.
J Biol Chem ; 297(4): 101196, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34529976

RESUMEN

Mitochondria undergo continuous cycles of fission and fusion to promote inheritance, regulate quality control, and mitigate organelle stress. More recently, this process of mitochondrial dynamics has been demonstrated to be highly sensitive to nutrient supply, ultimately conferring bioenergetic plasticity to the organelle. However, whether regulators of mitochondrial dynamics play a causative role in nutrient regulation remains unclear. In this study, we generated a cellular loss-of-function model for dynamin-related protein 1 (DRP1), the primary regulator of outer membrane mitochondrial fission. Loss of DRP1 (shDRP1) resulted in extensive ultrastructural and functional remodeling of mitochondria, characterized by pleomorphic enlargement, increased electron density of the matrix, and defective NADH and succinate oxidation. Despite increased mitochondrial size and volume, shDRP1 cells exhibited reduced cellular glucose uptake and mitochondrial fatty acid oxidation. Untargeted transcriptomic profiling revealed severe downregulation of genes required for cellular and mitochondrial calcium homeostasis, which was coupled to loss of ATP-stimulated calcium flux and impaired substrate oxidation stimulated by exogenous calcium. The insights obtained herein suggest that DRP1 regulates substrate oxidation by altering whole-cell and mitochondrial calcium dynamics. These findings are relevant to the targetability of mitochondrial fission and have clinical relevance in the identification of treatments for fission-related pathologies such as hereditary neuropathies, inborn errors in metabolism, cancer, and chronic diseases.


Asunto(s)
Señalización del Calcio , Dinaminas/metabolismo , Mitocondrias Musculares/metabolismo , Dinámicas Mitocondriales , Línea Celular , Dinaminas/genética , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Humanos , Mitocondrias Musculares/genética , Oxidación-Reducción
4.
Am J Physiol Renal Physiol ; 321(3): F356-F368, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34151592

RESUMEN

Low-protein (LP) diets extend lifespan through a comprehensive improvement in metabolic health across multiple tissues and organs. Many of these metabolic responses to protein restriction are secondary to transcriptional activation and release of FGF21 from the liver. However, the effects of an LP diet on the kidney in the context of aging has not been examined. Therefore, the goal of the current study was to investigate the impact of chronic consumption of an LP diet on the kidney in aging mice lacking FGF21. Wild-type (WT; C57BL/6J) and FGF21 knockout (KO) mice were fed a normal protein diet (20% casein) or an LP (5% casein) diet ad libitum from 3 to 22 mo of age. The LP diet led to a decrease in kidney weight and urinary albumin-to-creatinine ratio in both WT and FGF21 KO mice. Although the LP diet produced only mild fibrosis and infiltration of leukocytes in WT kidneys, the effects were significantly exacerbated by the absence of FGF21. Accordingly, transcriptomic analysis showed that inflammation-related pathways were significantly enriched and upregulated in response to LP diet in FGF21 KO mice but not WT mice. Collectively, these data demonstrate that the LP diet negatively affected the kidney during aging, but in the absence of FGF21, the LP diet-induced renal damage and inflammation were significantly worse, indicating a protective role of FGF21 in the kidney.NEW & NOTEWORTHY Long-term protein restriction is not advantageous for an otherwise healthy, aging kidney, as it facilitates the development of renal tubular injury and inflammatory cell infiltration. We provide evidence using FGF21 knockout animals that FGF21 is essential to counteract the renal injury and inflammation during aging on a low-protein diet.


Asunto(s)
Envejecimiento/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Inflamación/tratamiento farmacológico , Hígado/efectos de los fármacos , Animales , Dieta con Restricción de Proteínas , Factores de Crecimiento de Fibroblastos/metabolismo , Inflamación/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis/metabolismo
5.
Am J Physiol Renal Physiol ; 318(5): F1237-F1245, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32223308

RESUMEN

Changes in mitochondrial function are central to many forms of kidney disease, including acute injury, diabetic nephropathy, hypertension, and chronic kidney diseases. As such, there is an increasing need for reliable and fast methods for assessing mitochondrial respiratory function in renal cells. Despite being indispensable for many mechanistic studies, cultured cells or isolated mitochondria, however, often do not recapitulate in vivo or close to in vivo situations. Cultured and/or immortalized cells often change their bioenergetic profile and phenotype compared with in vivo or ex vivo situations, and isolated mitochondria are simply removed from their cellular milieu. This is especially important for extremely complex organs such as the kidney. Here, we report the development and validation of a new approach for the rapid assessment of mitochondrial oxygen consumption on freshly isolated glomeruli or proximal tubular fragments using Agilent SeaHorse XFe24 and XF96 Extracellular Flux Analyzers. We validated the technique in several healthy and diseased rodent models: the C57BL/6J mouse, the diabetic db/db mouse and matching db/+ control mouse, and the Dahl salt-sensitive rat. We compared the data to respiration from isolated mitochondria. The method can be adapted and used for the rapid assessment of mitochondrial oxygen consumption from any rodent model of the investigator's choice. The isolation methods presented here ensure viable and functional proximal tubular fragments and glomeruli, with a preserved cellular environment for studying mitochondrial function within the context of their surroundings and interactions.


Asunto(s)
Diabetes Mellitus/metabolismo , Metabolismo Energético , Hipertensión/metabolismo , Glomérulos Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Mitocondrias/metabolismo , Animales , Respiración de la Célula , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Femenino , Hipertensión/patología , Glomérulos Renales/patología , Túbulos Renales Proximales/patología , Masculino , Ratones Endogámicos C57BL , Mitocondrias/patología , Consumo de Oxígeno , Ratas Endogámicas Dahl
6.
Am J Physiol Renal Physiol ; 319(6): F1117-F1124, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33135479

RESUMEN

Sex differences (biological distinctions between males and females) present a complex interplay of genetic, developmental, biological, and environmental factors. More and more studies are shedding light on the importance of sex differences in normal physiology and susceptibility to cancer, cardiovascular and renal conditions, and neurodegenerative diseases. This mini-review is devoted to the role of sex dimorphisms in renal function, with a focus on the distinctions between male and female mitochondria. Here, we cover the aspects of renal mitochondrial bioenergetics where sex differences have been reported to date, for instance, biogenesis, reactive oxygen species production, and oxidative stress. Special attention is devoted to the effects of sex hormones, such as estrogen and testosterone, on mitochondrial bioenergetics in the kidney in physiology and pathophysiology.


Asunto(s)
Investigación Biomédica , Metabolismo Energético , Hormonas Esteroides Gonadales/metabolismo , Enfermedades Renales/metabolismo , Riñón/metabolismo , Mitocondrias/metabolismo , Nefrología , Animales , Femenino , Humanos , Riñón/fisiopatología , Enfermedades Renales/fisiopatología , Masculino , Caracteres Sexuales , Factores Sexuales
7.
J Lipid Res ; 59(12): 2321-2328, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30317185

RESUMEN

NADPH oxidase (NOX) enzymes are one of the major superoxide-generating systems in cells. NOX-generated superoxide has been suggested to promote insulin resistance in the liver. However, the role of NOX enzymes in mediating metabolic dysfunction in other insulin target tissues remains unclear. Here, we show that NOX3 expression is induced in differentiated 3T3-L1 adipocytes upon treatment with proinflammatory cytokines. Superoxide production increased concurrently with NOX3 protein expression in cytokine-treated adipocytes, which was inhibited by the NOX inhibitor diphenyleneiodonium (DPI). Treatment of adipocytes with cytokines increased lipolysis and decreased PPARγ activity. Interestingly, treatment with DPI blunted lipolysis activation by cytokines but failed to restore PPARγ activity. siRNA-mediated NOX3 downregulation also prevented cytokine-induced superoxide generation and lipolysis. In line with increasing lipolysis, cytokines increased the phosphorylation of hormone-sensitive lipase (HSL), which was reversed by treatment with DPI and silencing of NOX3 expression. We conclude that NOX3 is a cytokine-inducible superoxide-generating enzyme in adipocytes, which promotes lipolysis through increasing phosphorylation of HSL. This suggests a key role for NOX3-mediated superoxide production in the increased adipocyte lipolysis in inflammatory settings.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Citocinas/farmacología , Lipólisis/fisiología , NADPH Oxidasas/metabolismo , Superóxidos/metabolismo , Células 3T3-L1 , Animales , Inflamación/metabolismo , Resistencia a la Insulina , Lipólisis/efectos de los fármacos , Lipólisis/genética , Ratones , NADPH Oxidasas/genética , Compuestos Onio/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
8.
Am J Physiol Endocrinol Metab ; 309(8): E715-26, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26306596

RESUMEN

Proinflammatory cytokines impact islet ß-cell mass and function by altering the transcriptional activity within pancreatic ß-cells, producing increases in intracellular nitric oxide abundance and the synthesis and secretion of immunomodulatory proteins such as chemokines. Herein, we report that IL-1ß, a major mediator of inflammatory responses associated with diabetes development, coordinately and reciprocally regulates chemokine and insulin secretion. We discovered that NF-κB controls the increase in chemokine transcription and secretion as well as the decrease in both insulin secretion and proliferation in response to IL-1ß. Nitric oxide production, which is markedly elevated in pancreatic ß-cells exposed to IL-1ß, is a negative regulator of both glucose-stimulated insulin secretion and glucose-induced increases in intracellular calcium levels. By contrast, the IL-1ß-mediated production of the chemokines CCL2 and CCL20 was not influenced by either nitric oxide levels or glucose concentration. Instead, the synthesis and secretion of CCL2 and CCL20 in response to IL-1ß were dependent on NF-κB transcriptional activity. We conclude that IL-1ß-induced transcriptional reprogramming via NF-κB reciprocally regulates chemokine and insulin secretion while also negatively regulating ß-cell proliferation. These findings are consistent with NF-κB as a major regulatory node controlling inflammation-associated alterations in islet ß-cell function and mass.


Asunto(s)
Quimiocinas/metabolismo , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Interleucina-1beta/metabolismo , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Quimiocinas/genética , Espectroscopía de Resonancia por Spin del Electrón , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Immunoblotting , Insulina/genética , Secreción de Insulina , Insulinoma , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Consumo de Oxígeno , Neoplasias Pancreáticas , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Ratas Zucker , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Ribosómica S9 , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Células Tumorales Cultivadas
9.
Curr Diab Rep ; 15(7): 40, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25957525

RESUMEN

Although diabetes is mainly diagnosed based on elevated glucose levels, dyslipidemia is also observed in these patients. Chronic kidney disease (CKD), a frequent occurrence in patients with diabetes, is associated with major abnormalities in circulating lipoproteins and renal lipid metabolism. At baseline, most renal epithelial cells rely on fatty acids as their energy source. CKD, including that which occurs in diabetes, is characterized by tubule epithelial lipid accumulation. Whether this is due to increased uptake or greater local fatty acid synthesis is unknown. We have recently shown that CKD also leads to decreased fatty acid oxidation, which might be an additional mechanism leading to lipid accumulation. Defective fatty acid utilization causes energy depletion resulting in increased apoptosis and dedifferentiation, which in turn contributes to fibrosis and CKD progression. Enhanced fatty acid oxidation in the kidney induced by fenofibrate, a peroxisomal proliferator-activated receptor (PPAR)-α agonist, showed benefit in mouse models of CKD. Fenofibrate treatment also reduced albuminuria in patients with diabetes in multiple clinical trials. Taken together, these findings suggest that further understanding of lipid metabolism in diabetic kidney disease may lead to novel therapeutic approaches.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Metabolismo de los Lípidos , Animales , Metabolismo Energético , Células Epiteliales/metabolismo , Ácidos Grasos/metabolismo , Humanos , Receptores Activados del Proliferador del Peroxisoma/metabolismo
10.
Am J Physiol Renal Physiol ; 306(8): F896-906, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24500687

RESUMEN

Albuminuria is associated with metabolic syndrome and diabetes. It correlates with the progression of chronic kidney disease, particularly with tubular atrophy. The fatty acid load on albumin significantly increases in obesity, presenting a proinflammatory environment to the proximal tubules. However, little is known about changes in the redox milieu during fatty acid overload and how redox-sensitive mechanisms mediate cell death. Here, we show that albumin with fatty acid impurities or conjugated with palmitate but not albumin itself compromised mitochondrial and cell viability, membrane potential and respiration. Fatty acid overload led to a redox imbalance which deactivated the antioxidant protein peroxiredoxin 2 and caused a peroxide-mediated apoptosis through the redox-sensitive pJNK/caspase-3 pathway. Transfection of tubular cells with peroxiredoxin 2 was protective and mitigated apoptosis. Mitochondrial fatty acid entry and ceramide synthesis modulators suggested that mitochondrial ß oxidation but not ceramide synthesis may modulate lipotoxic effects on tubular cell survival. These results suggest that albumin overloaded with fatty acids but not albumin itself changes the redox environment in the tubules, inducing a peroxide-mediated redox-sensitive apoptosis. Thus, mitigating circulating fatty acid levels may be an important factor in both preserving redox balance and preventing tubular cell damage in proteinuric diseases.


Asunto(s)
Albúminas/metabolismo , Apoptosis/efectos de los fármacos , Ácidos Grasos/farmacología , Albúminas/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/patología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Oxidación-Reducción , Palmitatos/metabolismo , Peroxirredoxinas/metabolismo , Ratas , Albúmina Sérica Bovina/farmacología
11.
Compr Physiol ; 14(1): 5225-5242, 2023 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-38158371

RESUMEN

According to the Centers for Disease Control and Prevention, 1 in 2 U.S. adults have hypertension, and more than 1 in 7 chronic kidney disease. In fact, hypertension is the second leading cause of kidney failure in the United States; it is a complex disease characterized by, leading to, and caused by renal dysfunction. It is well-established that hypertensive renal damage is accompanied by mitochondrial damage and oxidative stress, which are differentially regulated and manifested along the nephron due to the diverse structure and functions of renal cells. This article provides a summary of the relevant knowledge of mitochondrial bioenergetics and metabolism, focuses on renal mitochondrial function, and discusses the evidence that has been accumulated regarding the role of epithelial mitochondrial bioenergetics in the development of renal tissue dysfunction in hypertension. © 2024 American Physiological Society. Compr Physiol 14:5225-5242, 2024.


Asunto(s)
Hipertensión Renal , Hipertensión , Humanos , Riñón/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Hipertensión Renal/metabolismo
12.
Trends Endocrinol Metab ; 34(11): 764-777, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37633800

RESUMEN

Lysosomes are cellular organelles that function to catabolize both extra- and intracellular cargo, act as a platform for nutrient sensing, and represent a core signaling node integrating bioenergetic cues to changes in cellular metabolism. Although lysosomal amino acid and lipid sensing in metabolism has been well characterized, lysosomal glucose sensing and the role of lysosomes in glucose metabolism is unrefined. This review will highlight the role of the lysosome in glucose metabolism with a focus on lysosomal glucose and glycogen sensing, glycophagy, and lysosomal glucose transport and how these processes impact autophagy and energy metabolism. Additionally, the role of lysosomal glucose metabolism in genetic and metabolic diseases will be briefly discussed.


Asunto(s)
Autofagia , Lisosomas , Humanos , Lisosomas/metabolismo , Glucógeno/metabolismo , Glucosa/metabolismo , Metabolismo Energético
13.
Nat Metab ; 5(4): 607-625, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37024752

RESUMEN

The lifetime risk of kidney disease in people with diabetes is 10-30%, implicating genetic predisposition in the cause of diabetic kidney disease (DKD). Here we identify an expression quantitative trait loci (QTLs) in the cis-acting regulatory region of the xanthine dehydrogenase, or xanthine oxidoreductase (Xor), a binding site for C/EBPß, to be associated with diabetes-induced podocyte loss in DKD in male mice. We examine mouse inbred strains that are susceptible (DBA/2J) and resistant (C57BL/6J) to DKD, as well as a panel of recombinant inbred BXD mice, to map QTLs. We also uncover promoter XOR orthologue variants in humans associated with high risk of DKD. We introduced the risk variant into the 5'-regulatory region of XOR in DKD-resistant mice, which resulted in increased Xor activity associated with podocyte depletion, albuminuria, oxidative stress and damage restricted to the glomerular endothelium, which increase further with type 1 diabetes, high-fat diet and ageing. Therefore, differential regulation of Xor contributes to phenotypic consequences with diabetes and ageing.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Humanos , Masculino , Ratones , Animales , Nefropatías Diabéticas/genética , Xantina Deshidrogenasa/genética , Xantina Deshidrogenasa/metabolismo , Predisposición Genética a la Enfermedad , Ratones Endogámicos DBA , Ratones Endogámicos C57BL
14.
Adv Exp Med Biol ; 771: 272-87, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23393685

RESUMEN

Oxidative stress and diabetes, both Type 1 and Type 2 as well as their related conditions have been extensively studied. As diabetes, obesity and metabolic syndrome have reached at epidemic levels, there is a huge need and effort to understand the detailed molecular mechanisms of the possible redox imbalance, underlying the cause of pathology and progression of the disease. These studies provide new insights at cellular and subcellular levels to design effective clinical interventions. This chapter is intended to emphasize the latest knowledge and current evidence on the role of oxidative stress in diabetes as well as to discuss some key questions that are currently under discussion.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hiperglucemia/metabolismo , Estrés Oxidativo/fisiología , Humanos
15.
Redox Biol ; 58: 102520, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36334379

RESUMEN

While it is generally accepted that oxidative stress impacts the diabetic kidney and contributes to pathogenesis, there is a substantial lack of knowledge about the molecular entity and anatomic location of a variety of reactive species. Here we provide a novel "oxidative stress map" of the diabetic kidney - the first of its kind, and identify specific, oxidized and other reactive lipids and their location. We used the db/db mouse model and Desorption Electrospray Ionization (DESI) mass spectrometry combined with heatmap image analysis. We analyzed a comprehensive array of phospholipid peroxide species in normal (db/m) and diabetic (db/db) kidneys using DESI imaging. Oxilipidomics heatmaps of the kidneys were generated focusing on phospholipids and their potential peroxidized products. We identified those lipids that undergo peroxidation in diabetic nephropathy. Several phospholipid peroxides and their spatial distribution were identified that were specific to the diabetic kidney, with significant enrichment in oxygenated phosphatidylethanolamines (PE) and lysophosphatidylethanolamine. Beyond qualitative and semi-quantitative information about the targets, the approach also reveals the anatomic location and the extent of lipid peroxide signal propagation across the kidney. Our approach provides novel, in-depth information of the location and molecular entity of reactive lipids in an organ with a very heterogeneous landscape. Many of these reactive lipids have been previously linked to programmed cell death mechanisms. Thus, the findings may be relevant to understand what impact phospholipid peroxidation has on cell and mitochondria membrane integrity and redox lipid signaling in diabetic nephropathy.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Ratones , Animales , Fosfolípidos/metabolismo , Nefropatías Diabéticas/metabolismo , Oxidación-Reducción , Espectrometría de Masa por Ionización de Electrospray/métodos , Riñón/metabolismo , Diabetes Mellitus/metabolismo
16.
ACS Chem Neurosci ; 13(2): 257-274, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-34990116

RESUMEN

Post-traumatic stress disorder (PTSD) is associated with cognitive deficits, oxidative stress, and inflammation. Animal models have recapitulated features of PTSD, but no comparative RNA sequencing analysis of differentially expressed genes (DEGs) in the brain between PTSD and animal models of traumatic stress has been carried out. We compared DEGs from the prefrontal cortex (PFC) of an established stress model to DEGs from the dorsolateral PFC (dlPFC) of humans. We observed a significant enrichment of rat DEGs in human PTSD and identified 20 overlapping DEGs, of which 17 (85%) are directionally concordant. N,N-dimethyltryptamine (DMT) is a known indirect antioxidant, anti-inflammatory, and neuroprotective compound with antidepressant and plasticity-facilitating effects. We tested the capacity of DMT, the monoamine oxidase inhibitor (MAOI) harmaline, and "pharmahuasca" (DMT + harmaline) to reduce reactive oxygen species (ROS) production and inflammatory gene expression and to modulate neuroplasticity-related gene expression in the model. We administered DMT (2 mg/kg IP), harmaline (1.5 mg/kg IP), pharmahuasca, or vehicle every other day for 5 days, following a 30 day stress regiment. We measured ROS production in the PFC and hippocampus (HC) by electron paramagnetic resonance spectroscopy and sequenced total mRNA in the PFC. We also performed in vitro assays to measure the affinity and efficacy of DMT and harmaline at 5HT2AR compared to 5-HT. DMT and pharmahuasca reduced ROS production in the PFC and HC, while harmaline had mixed effects. Treatments normalized 9, 12, and 14 overlapping DEGs, and pathway analysis implicated that genes were involved in ROS production, inflammation, growth factor signaling, neurotransmission, and neuroplasticity.


Asunto(s)
N,N-Dimetiltriptamina , Trastornos por Estrés Postraumático , Animales , Corteza Prefontal Dorsolateral , Humanos , Ratas , Especies Reactivas de Oxígeno , Trastornos por Estrés Postraumático/tratamiento farmacológico , Estrés Psicológico/tratamiento farmacológico
17.
JCI Insight ; 7(7)2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35230975

RESUMEN

The importance of healthy mitochondrial function is implicated in the prevention of chronic kidney disease (CKD) and diabetic kidney disease (DKD). Sex differences also play important roles in DKD. Our previous studies revealed that mitochondrial substrate overload (modeled by homozygous deletion of carnitine acetyl-transferase [CrAT]) in proximal tubules causes renal injury. Here, we demonstrate the importance of intact mitochondrial substrate efflux by titrating the amount of overload through the generation of a heterozygous CrAT-KO model (PT-CrATHET mouse). Intriguingly, these animals developed renal injury similarly to their homozygous counterparts. Mitochondria were structurally and functionally impaired in both sexes. Transcriptomic analyses, however, revealed striking sex differences. Male mice shut down fatty acid oxidation and several other metabolism-related pathways. Female mice had a significantly weaker transcriptional response in metabolism, but activation of inflammatory pathways was prominent. Proximal tubular cells from PT-CrATHET mice of both sexes exhibited a shift toward a more glycolytic phenotype, but female mice were still able to oxidize fatty acid-based substrates. Our results demonstrate that maintaining mitochondrial substrate metabolism balance is crucial to satisfying proximal tubular energy demand. Our findings have potentially broad implications, as both the glycolytic shift and the sexual dimorphisms discovered herein offer potentially new modalities for future interventions for treating kidney disease.


Asunto(s)
Nefropatías Diabéticas , Mitocondrias , Animales , Nefropatías Diabéticas/metabolismo , Ácidos Grasos/metabolismo , Femenino , Homocigoto , Masculino , Ratones , Mitocondrias/metabolismo , Eliminación de Secuencia
18.
Am J Physiol Endocrinol Metab ; 300(6): E1047-58, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21386058

RESUMEN

Obesity and metabolic syndrome are associated with an increased risk for several diabetic complications, including diabetic nephropathy and chronic kidney diseases. Oxidative stress and mitochondrial dysfunction are often proposed mechanisms in various organs in obesity models, but limited data are available on the kidney. Here, we fed a lard-based high-fat diet to mice to investigate structural changes, cellular and subcellular oxidative stress and redox status, and mitochondrial biogenesis and function in the kidney. The diet induced characteristic changes, including glomerular hypertrophy, fibrosis, and interstitial scarring, which were accompanied by a proinflammatory transition. We demonstrate evidence for oxidative stress in the kidney through 3-nitrotyrosine and protein radical formation on high-fat diet with a contribution from iNOS and NOX-4 as well as increased generation of mitochondrial oxidants on carbohydrate- and lipid-based substrates. The increased H(2)O(2) emission in the mitochondria suggests altered redox balance and mitochondrial ROS generation, contributing to the overall oxidative stress. No major derailments were observed in respiratory function or biogenesis, indicating preserved and initially improved bioenergetic parameters and energy production. We suggest that, regardless of the oxidative stress events, the kidney developed an adaptation to maintain normal respiratory function as a possible response to an increased lipid overload. These findings provide new insights into the complex role of oxidative stress and mitochondrial redox status in the pathogenesis of the kidney in obesity and indicate that early oxidative stress-related changes, but not mitochondrial bioenergetic dysfunction, may contribute to the pathogenesis and development of obesity-linked chronic kidney diseases.


Asunto(s)
Dieta , Grasas de la Dieta/farmacología , Metabolismo Energético/efectos de los fármacos , Riñón/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Adaptación Fisiológica/fisiología , Animales , Ingestión de Energía/fisiología , Radicales Libres/metabolismo , Peróxido de Hidrógeno/metabolismo , Inmunohistoquímica , Inflamación/genética , Inflamación/metabolismo , Riñón/efectos de los fármacos , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Mitocondrias/efectos de los fármacos , NADPH Oxidasa 4 , NADPH Oxidasas/biosíntesis , NADPH Oxidasas/genética , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Superóxido Dismutasa/metabolismo , Tirosina/análogos & derivados , Tirosina/farmacología
19.
J Immunol ; 183(6): 4055-66, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19717511

RESUMEN

LPS-induced sepsis results in oxidative modification and inactivation of carboxypeptidase B1 (CPB1). In this study, immunoprecipitated CPB1 was probed for tyrosine nitration using monoclonal nitrotyrosine-specific Abs in a murine model of LPS-induced sepsis. Tyrosine nitration of CPB1 was significantly reduced in the presence of NO synthase (NOS) inhibitors and the xanthine oxidase (XO) inhibitor allopurinol and in NOS-3 knockout (KO) mice. CPB1 tyrosine nitration and loss of activity by the concerted action of NOS-3 and XO were also confirmed in vitro using both the NO donor 3-morpholinosydnonimine and peroxynitrite. Liquid chromatography/tandem mass spectrometry data indicated five sites of tyrosine nitration in vitro including Tyr(248), the tyrosine at the catalytic site. The site- and protein-specific nitration of CPB1 and the possible high nitration yield to inactivate it were elucidated by confocal microscopy. The studies indicated that CPB1 colocalized with NOS-3 in the cytosol of sinus-lining cells in the red pulp of the spleen. Further analysis of CPB1-immunoprecipitated samples indicated immunoreactivity to a monoclonal NOS-3 Ab, suggesting protein complex formation with CPB1. XO and NOS inhibitors and NOS-3 KO mice injected with LPS had decreased levels of C5a in spleens of septic mice, indicating peroxynitrite as a possible cause for CPB1 functional alteration. Thus, CPB1 colocalization, coupling, and proximity to NOS-3 in the sinus-lining cells of spleen red pulp could explain the site-specific tyrosine nitration and inactivation of CPB1. These results open up new avenues for the investigation of several enzymes involved in inflammation and their site-specific oxidative modifications by protein-protein interactions as well as their role in sepsis.


Asunto(s)
Carboxipeptidasa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Sepsis/metabolismo , Animales , Sitios de Unión , Carboxipeptidasa B/análisis , Lipopolisacáridos , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/análisis , Óxido Nítrico Sintasa de Tipo III/deficiencia , Nitrosación , Unión Proteica , Sepsis/inducido químicamente , Sepsis/enzimología , Bazo/química , Espectrometría de Masas en Tándem , Tirosina/metabolismo
20.
Proc Natl Acad Sci U S A ; 105(25): 8569-74, 2008 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-18562300

RESUMEN

The physiological effects of nitroglycerin as a potent vasodilator have long been documented. However, the molecular mechanisms by which nitroglycerin exerts its biological functions are still a matter of intense debate. Enzymatic pathways converting nitroglycerin to vasoactive compounds have been identified, but none of them seems to fully account for the reported clinical observations. Here, we demonstrate that nitroglycerin triggers constitutive nitric oxide synthase (NOS) activation, which is a major source of NO responsible for low-dose (1-10 nM) nitroglycerin-induced vasorelaxation. Our studies in cell cultures, isolated vessels, and whole animals identified endothelial NOS activation as a fundamental requirement for nitroglycerin action at pharmacologically relevant concentrations in WT animals.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitroglicerina/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Células Cultivadas , Endotelio Vascular/enzimología , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Ratones , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Fosforilación , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA