Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(21)2020 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-33121167

RESUMEN

Chronic kidney disease (CKD) is prevalent in 9.1% of the global population and is a significant public health problem associated with increased morbidity and mortality. CKD is associated with highly prevalent physiological and metabolic disturbances such as hypertension, obesity, insulin resistance, cardiovascular disease, and aging, which are also risk factors for CKD pathogenesis and progression. Podocytes and proximal tubular cells of the kidney strongly express AMP-activated protein kinase (AMPK). AMPK plays essential roles in glucose and lipid metabolism, cell survival, growth, and inflammation. Thus, metabolic disease-induced renal diseases like obesity-related and diabetic chronic kidney disease demonstrate dysregulated AMPK in the kidney. Activating AMPK ameliorates the pathological and phenotypical features of both diseases. As a metabolic sensor, AMPK regulates active tubular transport and helps renal cells to survive low energy states. AMPK also exerts a key role in mitochondrial homeostasis and is known to regulate autophagy in mammalian cells. While the nutrient-sensing role of AMPK is critical in determining the fate of renal cells, the role of AMPK in kidney autophagy and mitochondrial quality control leading to pathology in metabolic disease-related CKD is not very clear and needs further investigation. This review highlights the crucial role of AMPK in renal cell dysfunction associated with metabolic diseases and aims to expand therapeutic strategies by understanding the molecular and cellular processes underlying CKD.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Enfermedades Metabólicas/complicaciones , Insuficiencia Renal Crónica/metabolismo , Progresión de la Enfermedad , Regulación de la Expresión Génica , Humanos , Metabolismo de los Lípidos , Enfermedades Metabólicas/metabolismo , Mitocondrias/metabolismo , Insuficiencia Renal Crónica/etiología
2.
Int J Mol Sci ; 22(1)2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396267

RESUMEN

Exercise training is now recognized as an interesting therapeutic strategy in managing obesity and its related disorders. However, there is still a lack of knowledge about its impact on obesity-induced chronic kidney disease (CKD). Here, we investigated the effects of a delayed protocol of endurance exercise training (EET) as well as the underlying mechanism in obese mice presenting CKD. Mice fed a high-fat diet (HFD) or a low-fat diet (LFD) for 12 weeks were subsequently submitted to an 8-weeks EET protocol. Delayed treatment with EET in obese mice prevented body weight gain associated with a reduced calorie intake. EET intervention counteracted obesity-related disorders including glucose intolerance, insulin resistance, dyslipidaemia and hepatic steatosis. Moreover, our data demonstrated for the first time the beneficial effects of EET on obesity-induced CKD as evidenced by an improvement of obesity-related glomerulopathy, tubulo-interstitial fibrosis, inflammation and oxidative stress. EET also prevented renal lipid depositions in the proximal tubule. These results were associated with an improvement of the AMPK pathway by EET in renal tissue. AMPK-mediated phosphorylation of ACC and ULK-1 were particularly enhanced leading to increased fatty acid oxidation and autophagy improvement with EET in obese mice.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Dieta Alta en Grasa/efectos adversos , Obesidad/complicaciones , Condicionamiento Físico Animal , Insuficiencia Renal Crónica/prevención & control , Proteínas Quinasas Activadas por AMP/genética , Animales , Intolerancia a la Glucosa , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Fosforilación , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología
3.
Arterioscler Thromb Vasc Biol ; 38(7): 1427-1439, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880486

RESUMEN

The endothelial glycocalyx (EG), which covers the apical surface of the endothelial cells and floats into the lumen of the vessels, is a key player in vascular integrity and cardiovascular homeostasis. The EG is composed of PGs (proteoglycans), glycoproteins, glycolipids, and glycosaminoglycans, in particular hyaluronan (HA). HA seems to be implicated in most of the functions described for EG such as creating a space between blood and the endothelium, controlling vessel permeability, restricting leukocyte and platelet adhesion, and allowing an appropriate endothelial response to flow variation through mechanosensing. The amount of HA in the EG may be regulated by HYAL (hyaluronidase) 1, the most active somatic hyaluronidase. HYAL1 seems enriched in endothelial cells through endocytosis from the bloodstream. The role of the other main somatic hyaluronidase, HYAL2, in the EG is uncertain. Damage to the EG, accompanied by shedding of one or more of its components, is an early sign of various pathologies including diabetes mellitus. Shedding increases the blood or plasma concentration of several EG components, such as HA, heparan sulfate, and syndecan. The plasma levels of these molecules can then be used as sensitive markers of EG degradation. This has been shown in type 1 and type 2 diabetic patients. Recent experimental studies suggest that preserving the size and amount of EG HA in the face of diabetic insults could be a useful novel therapeutic strategy to slow diabetic complications. One way to achieve this goal, as suggested by a murine model of HYAL1 deficiency, may be to inhibit the function of HYAL1. The same approach may succeed in other pathological situations involving endothelial dysfunction and EG damage.


Asunto(s)
Diabetes Mellitus/enzimología , Angiopatías Diabéticas/enzimología , Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Glicocálix/enzimología , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/metabolismo , Animales , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/fisiopatología , Angiopatías Diabéticas/diagnóstico , Angiopatías Diabéticas/fisiopatología , Angiopatías Diabéticas/prevención & control , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Inhibidores Enzimáticos/uso terapéutico , Glicocálix/efectos de los fármacos , Glicocálix/patología , Humanos , Hialuronoglucosaminidasa/antagonistas & inhibidores , Mecanotransducción Celular
4.
Am J Physiol Heart Circ Physiol ; 314(2): H350-H358, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29101168

RESUMEN

Angiotensin I-converting enzyme (ACE) levels in humans are under strong genetic influence. Genetic variation in ACE has been linked to risk for and progression of cardiovascular and renal diseases. Causality has been documented in genetically modified mice, but the mechanisms underlying causality are not completely elucidated. To further document the vascular and renal consequences of a moderate genetic increase in ACE synthesis, we studied genetically modified mice carrying three copies of the ACE gene (three-copy mice) and littermate wild-type animals (two-copy mice). We investigated peripheral and renal vascular reactivity to angiotensin II and bradykinin in vivo by measuring blood pressure and renal blood flow after intravenous administration and also reactivity of isolated glomerular arterioles by following intracellular Ca2+ mobilization. Carrying three copies of the ACE gene potentiated the systemic and renal vascular responses to angiotensin II over the whole range of peptide concentration tested. Consistently, the response of isolated glomerular afferent arterioles to angiotensin II was enhanced in three-copy mice. In these mice, signaling pathways triggered by endothelial activation by bradykinin or carbachol in glomerular arterioles were also altered. Although the nitric oxide (NO) synthase (NOS)/NO pathway was not functional in arterioles of two-copy mice, in muscular efferent arterioles of three-copy mice NOS3 gene expression was induced and NO mediated the effect of bradykinin or carbachol. These data document new and unexpected vascular consequences of a genetic increase in ACE synthesis. Enhanced vasoconstrictor effect of angiotensin II may contribute to the risk for cardiovascular and renal diseases linked to genetically high ACE levels. NEW & NOTEWORTHY A moderate genetic increase in angiotensin I-converting enzyme (ACE) in mice similar to the effect of the ACE gene D allele in humans unexpectedly potentiates the systemic and renal vasoconstrictor responses to angiotensin II. It also alters the endothelial signaling pathways triggered by bradykinin or carbachol in glomerular efferent arterioles.


Asunto(s)
Angiotensina II/farmacología , Presión Arterial/efectos de los fármacos , Arteriolas/efectos de los fármacos , Bradiquinina/farmacología , Glomérulos Renales/irrigación sanguínea , Peptidil-Dipeptidasa A/biosíntesis , Circulación Renal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatadores/farmacología , Animales , Arteriolas/enzimología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Carbacol/farmacología , Inducción Enzimática , Femenino , Genotipo , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Peptidil-Dipeptidasa A/genética , Fenotipo
5.
Exp Physiol ; 103(1): 125-140, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28944982

RESUMEN

NEW FINDINGS: What is the central question of this study? The metabolic pathways regulating the effects of obesity on the kidney remain unknown. We sought to determine whether inducible nitric oxide synthase (iNOS) is involved in the underlying mechanisms of high-fat diet-induced kidney disease using a specific iNOS inhibitor, N6-(1-iminoethyl)-l-lysine hydrochloride (L-NIL). What is the main finding and its importance? We did not demonstrate an upregulation of iNOS renal expression after high caloric intake, suggesting that iNOS might not be a crucial player in the development of obesity-induced kidney disease. Although L-NIL treatment clearly ameliorated systemic metabolic parameters, the effect on loss of renal function, impairment of tubular integrity, oxidative stress and inflammation appeared to be more moderate. Central obesity is related to caloric excess, promoting deleterious cellular responses in targeted organs. Nitric oxide (NO) has been determined as a key player in the pathogenesis of metabolic diseases. Here, we investigated the implication of inducible NO synthase (iNOS) in the development of obesity-induced kidney disease. C57Bl/6 male mice were randomized to a low-fat diet (LFD) or a high-fat diet (HFD) and treated with N6-(1-iminoethyl)-l-lysine hydrochloride (L-NIL), a specific iNOS inhibitor, for 16 weeks. Mice fed an HFD exhibited a significant increase in body weight, fasting blood glucose and plasma concentrations of non-esterified fatty acids, triglyceride and insulin. Inhibition of iNOS prevented these changes in mice fed an HFD. Interestingly, the significant increase in albuminuria and mesangial matrix expansion were not ameliorated with L-NIL, whereas a significant decrease in proteinuria, N-acetyl-ß-d-glucosaminidase excretion and renal triglyceride content were found, suggesting that iNOS inhibition is more suitable for tubular function than glomerular function. The urinary concentration of hydrogen peroxide, a stable product of reactive oxygen species production, that was found to be increased in mice fed an HFD, was significantly reduced with L-NIL. Finally, despite a moderate effect of L-NIL on inflammatory processes in the kidney, we demonstrated a positive impact of this treatment on adipocyte hypertrophy and on adipose tissue inflammation. These results suggest that inhibition of iNOS leads to a moderate beneficial effect on kidney function in mice fed an HFD. Further studies are needed for better understanding of the role of iNOS in obesity-induced kidney disease.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Enfermedades Renales/enzimología , Enfermedades Renales/patología , Riñón/patología , Riñón/fisiología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Animales , Inhibidores Enzimáticos/farmacología , Riñón/efectos de los fármacos , Enfermedades Renales/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/fisiología
6.
Int J Mol Sci ; 18(2)2017 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-28146082

RESUMEN

The term "aristolochic acid nephropathy" (AAN) is used to include any form of toxic interstitial nephropathy that is caused either by ingestion of plants containing aristolochic acids (AA) as part of traditional phytotherapies (formerly known as "Chinese herbs nephropathy"), or by the environmental contaminants in food (Balkan endemic nephropathy). It is frequently associated with urothelial malignancies. Although products containing AA have been banned in most of countries, AAN cases remain regularly reported all over the world. Moreover, AAN incidence is probably highly underestimated given the presence of AA in traditional herbal remedies worldwide and the weak awareness of the disease. During these two past decades, animal models for AAN have been developed to investigate underlying molecular and cellular mechanisms involved in AAN pathogenesis. Indeed, a more-in-depth understanding of these processes is essential to develop therapeutic strategies aimed to reduce the global and underestimated burden of this disease. In this regard, our purpose was to build a broad overview of what is currently known about AAN. To achieve this goal, we aimed to summarize the latest data available about underlying pathophysiological mechanisms leading to AAN development with a particular emphasis on the imbalance between vasoactive factors as well as a focus on the vascular events often not considered in AAN.


Asunto(s)
Ácidos Aristolóquicos/efectos adversos , Medicamentos Herbarios Chinos/efectos adversos , Nefritis Intersticial/etiología , Animales , Ácidos Aristolóquicos/química , Ácidos Aristolóquicos/metabolismo , Nefropatía de los Balcanes/diagnóstico , Nefropatía de los Balcanes/epidemiología , Nefropatía de los Balcanes/etiología , Biopsia , Transformación Celular Neoplásica/inducido químicamente , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/metabolismo , Fibrosis , Humanos , Neoplasias Renales/etiología , Nefritis Intersticial/diagnóstico , Nefritis Intersticial/epidemiología , Estrés Oxidativo
7.
Exp Physiol ; 101(1): 193-206, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26442795

RESUMEN

Aristolochic acid (AA) nephropathy (AAN), a progressive tubulointerstitial injury of toxic origin, is characterized by early and transient acute tubular necrosis. This process has been demonstrated to be associated with reduced nitric oxide (NO) production, which can disrupt the regulation of renal function. In this study, we tested the hypothesis that L-arginine (L-Arg) supplementation could restore renal function and reduce renal injury after AA intoxication. C57BL/6 J male mice were randomly subjected to daily i.p. injection of either sterile saline solution or AA (2.5 mg kg(-1)) for 4 days. To determine whether AA-induced renal injuries were linked to reduced NO production, L-Arg, a substrate for NO synthase, was supplemented (5%) in drinking water. Mice intoxicated with AA exhibited features of rapid-onset acute kidney injury, including polyuria, significantly increased plasma creatinine concentrations, proteinuria and fractional excretion of sodium (P < 0.05), along with severe proximal tubular cell injury and increased NADPH oxidase 2 (Nox2)-derived oxidative stress (P < 0.05). This was associated with a significant reduction in NO bioavailability. L-Arg supplementation in AA-treated mice significantly increased NO bioavailability, which in turn improved renal function (creatininaemia, polyuria, proteinuria, fractional excreted sodium and N-acetyl-ß-D-glucosaminidase enzymuria) and renal structure (tubular necrosis and tubular cell apoptosis). These changes were associated with significant reductions in Nox2 expression and in production of reactive oxygen species and with an increase in antioxidant concentrations. Our results demonstrate that preservation of NO bioavailability leads to renal protection in AA-induced acute kidney injury by reducing oxidative stress and maintaining renal function.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Ácidos Aristolóquicos , Óxido Nítrico/uso terapéutico , Lesión Renal Aguda/patología , Animales , Arginina/farmacología , Creatinina/sangre , GMP Cíclico/orina , Riñón/patología , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Poliuria/inducido químicamente , Poliuria/prevención & control , Proteinuria/inducido químicamente , Proteinuria/prevención & control , Sodio/orina , Superóxido Dismutasa/metabolismo
8.
Kidney Int ; 88(1): 61-71, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25715119

RESUMEN

Renal ischemia-reperfusion injury (IRI) is a pathological process that may lead to acute renal failure and chronic dysfunction in renal allografts. During IRI, hyaluronan (HA) accumulates in the kidney, but suppression of HA accumulation during IRI protects the kidney from ischemic insults. Here we tested whether Hyal1-/- and Hyal2-/- mice display exacerbated renal damage following unilateral IRI due to a higher HA accumulation in the post-ischemic kidney compared with that in the kidney of wild-type mice. Two days after IRI in male mice there was accumulation of HA and CD44 in the kidney, marked tubular damage, infiltration, and increase creatininemia in wild-type mice. Knockout mice exhibited higher amounts of HA and higher creatininemia. Seven days after injury, wild-type mice had a significant decrease in renal damage, but knockout mice still displayed exacerbated inflammation. HA and CD44 together with α-smooth muscle actin and collagen types I and III expression were increased in knockout compared with wild-type mice 30 days after IRI. Thus, both HA-degrading enzymes seem to be protective against IRI most likely by reducing HA accumulation in the post-ischemic kidney and decreasing the inflammatory processes. Deficiency in either HYAL1 or HYAL2 leads to enhanced HA accumulation in the post-ischemic kidney and consequently worsened inflammatory response, increased tubular damage, and fibrosis.


Asunto(s)
Lesión Renal Aguda/etiología , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/deficiencia , Riñón/patología , Mucopolisacaridosis/complicaciones , Daño por Reperfusión/complicaciones , Actinas/metabolismo , Lesión Renal Aguda/genética , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL2/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Creatinina/sangre , Fibrosis , Proteínas Ligadas a GPI/genética , Receptores de Hialuranos/metabolismo , Hialuronoglucosaminidasa/genética , Túbulos Renales/patología , Recuento de Leucocitos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mucopolisacaridosis/genética , Nefritis/etiología , Nefritis/genética , Nefritis/patología , Neutrófilos , ARN Mensajero/metabolismo , Daño por Reperfusión/metabolismo
9.
Clin Exp Pharmacol Physiol ; 41(11): 911-20, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25115485

RESUMEN

The Wistar-Furth (WF) rat strain is usually used in models of full major histocompatibility complex-mismatched kidney transplantation. Because these rats have been demonstrated to be resistant to several models of chronic kidney disease, the aim of the present study was to investigate their potential resistance to renal ischaemia-reperfusion (I/R) injury compared with another strain, namely Wistar-Hanover (WH) rats. Anaesthetized male WH and WF rats were submitted to I/R by occlusion of the left renal artery and contralateral nephrectomy. Urine, blood and tissue samples were collected at different time points after I/R to evaluate renal function, inflammation and tubular injury, along with determination of nitric oxide synthase (NOS) expression and thromboxane A2 (TxA2 ) production. Post-ischaemic renal function was better preserved in WF than WH rats, as evidenced by reduced levels of creatininaemia, urinary neutrophil gelatinase-associated lipocalin excretion and proteinuria. In addition, WF rats had less intrarenal inflammation than WH rats after I/R injury. These observations were associated with maintenance of neuronal NOS expression, along with lower induction of inducible NOS expression in WF versus WH rats. Moreover, WF rats excreted a significantly lower amount of TxB2 . The results indicate that WF rats are more resistant to an I/R injury than WH rats in terms of renal function and inflammation. These observations are associated with differential regulation of intrarenal NOS expression, as well as a reduction in thromboxane production, which could contribute to a better outcome for the postischaemic kidney in WF rats.


Asunto(s)
Modelos Animales de Enfermedad , Riñón/metabolismo , Óxido Nítrico/biosíntesis , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Tromboxano A2/biosíntesis , Enfermedad Aguda , Animales , Dinoprostona/orina , Riñón/irrigación sanguínea , Riñón/inmunología , Pruebas de Función Renal , Masculino , Óxido Nítrico Sintasa/genética , Estrés Oxidativo , Ratas Endogámicas WF , Reacción en Cadena en Tiempo Real de la Polimerasa , Daño por Reperfusión/inmunología , Daño por Reperfusión/orina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tromboxano B2/orina
10.
Nephrol Dial Transplant ; 28(10): 2484-93, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24078641

RESUMEN

BACKGROUND: Ischaemia-reperfusion injury (IRI) to the kidney is a complex pathophysiological process that leads to acute renal failure and chronic dysfunction in renal allografts. It was previously demonstrated that during IRI, hyaluronan (HA) accumulates in the cortical and external medullary interstitium along with an increased expression of its main receptor, CD44, on inflammatory and tubular cells. The HA-CD44 pair may be involved in persistent post-ischaemic inflammation. Thus, we sought to determine the role of HA in the pathophysiology of ischaemia-reperfusion (IR) by preventing its accumulation in post-ischaemic kidney. METHODS: C57BL/6 mice received a diet containing 4-methylumbelliferone (4-MU), a potent HA synthesis inhibitor. At the end of the treatment, unilateral renal IR was induced and mice were euthanized 48 h or 30 days post-IR. RESULTS: 4-MU treatment for 14 weeks reduced the plasma HA level and intra-renal HA content at 48 h post-IR, as well as CD44 expression, creatininemia and histopathological lesions. Moreover, inflammation was significantly attenuated and proliferation was reduced in animals treated with 4-MU. In addition, 4-MU-treated mice had a significantly reduced expression of α-SMA and collagen types I and III, i.e. less renal fibrosis, 30 days after IR compared with untreated mice. CONCLUSION: Our results demonstrate that HA plays a significant role in the pathogenesis of IRI, perhaps in part through reduced expression of CD44. The suppression of HA accumulation during IR may protect renal function against ischaemic insults.


Asunto(s)
Lesión Renal Aguda/prevención & control , Modelos Animales de Enfermedad , Ácido Hialurónico/antagonistas & inhibidores , Himecromona/farmacología , Inflamación/prevención & control , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/etiología , Animales , Ácido Hialurónico/metabolismo , Indicadores y Reactivos/farmacología , Inflamación/etiología , Pruebas de Función Renal , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/complicaciones
11.
Biol Sex Differ ; 14(1): 63, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37770988

RESUMEN

BACKGROUND: Sex differences have been observed in the development of obesity-related complications in patients, as well as in animal models. Accumulating evidence suggests that sex-dependent regulation of lipid metabolism contributes to sex-specific physiopathology. Lipid accumulation in the renal tissue has been shown to play a major role in the pathogenesis of obesity-induced kidney injury. Unlike in males, the physiopathology of the disease has been poorly described in females, particularly regarding the lipid metabolism adaptation. METHODS: Here, we compared the lipid profile changes in the kidneys of female and male mice fed a high-fat diet (HFD) or low-fat diet (LFD) by lipidomics and correlated them with pathophysiological changes. RESULTS: We showed that HFD-fed female mice were protected from insulin resistance and hepatic steatosis compared to males, despite similar body weight gains. Females were particularly protected from renal dysfunction, oxidative stress, and tubular lipid accumulation. Both HFD-fed male and female mice presented dyslipidemia, but lipidomic analysis highlighted differential renal lipid profiles. While both sexes presented similar neutral lipid accumulation with obesity, only males showed increased levels of ceramides and phospholipids. Remarkably, protection against renal lipotoxicity in females was associated with enhanced renal adiponectin and AMP-activated protein kinase (AMPK) signaling. Circulating adiponectin and its renal receptor levels were significantly lower in obese males, but were maintained in females. This observation correlated with the maintained basal AMPK activity in obese female mice compared to males. CONCLUSIONS: Collectively, our findings suggest that female mice are protected from obesity-induced renal dysfunction and lipotoxicity associated with enhanced adiponectin and AMPK signaling compared to males.


Obesity-related complications can differ between men and women due to sex-specific differences in how fats are handled. Here, we studied the effects of high-fat diet on the kidneys of male and female mice. We found that despite gaining similar weight, obese female mice were better protected against insulin resistance, liver fat accumulation, and kidney damage caused by obesity than males. In particular, female mice were protected against lipid accumulation in the kidneys. We further analyzed the lipid profile in the kidneys of both male and female mice and observed differences in the amount and nature of the accumulated lipids. Male mice had increased levels of specific lipids, which may contribute to their higher risk of kidney damage. In contrast, female mice showed better lipid metabolism adaptation, which helped to protect their kidneys. This study also revealed an association between higher levels of the protein hormone adiponectin and higher activity of the cellular energy master regulator protein AMPK in obese females. These proteins may help prevent obesity-induced kidney damage. In obese males, these protective proteins are reduced and are associated with kidney damage. In conclusion, this study suggests that female mice are naturally shielded from obesity-induced kidney damage and lipid accumulation in the kidneys. Obesity in males is associated with the presence of potentially toxic lipids and dysregulated renal metabolism. Understanding these sex-related differences in obesity-related complications could lead to better management and treatment of kidney problems in both men and women.


Asunto(s)
Adiponectina , Enfermedades Renales , Animales , Femenino , Masculino , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Riñón/metabolismo , Enfermedades Renales/etiología , Lipidómica , Lípidos , Obesidad/metabolismo , Caracteres Sexuales
12.
Nephrol Dial Transplant ; 27(10): 3771-81, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22529164

RESUMEN

BACKGROUND: The turnover of hyaluronan (HA), especially the production of low-molecular-weight fragments of HA, was examined in a model of unilateral renal ischaemia-reperfusion (IR) in rats. METHODS: HA was extracted from the outer and inner stripe of the outer medulla (OSOM and ISOM) at different times following IR. Its fragmentation was measured using membrane filtration and size-exclusion chromatography. Quantitative reverse transcription-polymerase chain reaction, zymography and immunohistochemistry were used to assess the expression and localization of various forms of HA synthase (HAS) and hyaluronidase (HYAL). Macrophage infiltration was evaluated using immunohistochemistry. RESULTS: HA accumulated at Day 1 mostly as high-molecular-weight (HMW) species with an elution profile similar to a reference 2500 kDa HA and at Day 14 mostly as medium- to low-size fragments. Within 1 day, HAS1 messenger RNA was up-regulated > 50- and 35-fold in OSOM and ISOM, respectively. Thereafter, HAS1 tended to normalize, while HAS2 increased steadily. Both synthetic enzymes were localized around tubules and in the interstitium. Conversely, HYAL1, HYAL2 and global hyaluronidase activity were repressed during the first 24 h. The patterns were identical in the OSOM and ISOM despite markedly different amounts of HA at baseline. There was no obvious correlation between HA deposits and macrophage infiltration. CONCLUSIONS: In the post-ischaemic kidney, HA starts to accumulate at Day 1 mostly as HMW species. Later on, a large proportion becomes degraded into smaller fragments. This pattern is explained by coordinated changes in the expression of HA synthases and hyaluronidases, especially an early induction of HAS1. The current data open the door to timed pharmacological interventions blocking the production of HA fragments.


Asunto(s)
Ácido Hialurónico/biosíntesis , Isquemia/metabolismo , Riñón/irrigación sanguínea , Riñón/metabolismo , Animales , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Hialuronano Sintasas , Ácido Hialurónico/química , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/genética , Hialuronoglucosaminidasa/metabolismo , Isquemia/genética , Riñón/lesiones , Riñón/patología , Médula Renal/metabolismo , Macrófagos/patología , Masculino , Peso Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo
13.
Front Med (Lausanne) ; 9: 822870, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35602498

RESUMEN

Aristolochic acid nephropathy (AAN) is a progressive tubulointerstitial nephritis caused by the intake of aristolochic acids (AA) contained in Chinese herbal remedies or contaminated food. AAN is characterized by tubular atrophy and interstitial fibrosis, characterizing advanced kidney disease. It is established that sustained or recurrent acute kidney injury (AKI) episodes contribute to the progression of CKD. Therefore, the study of underlying mechanisms of AA-induced nephrotoxicity could be useful in understanding the complex AKI-to-CKD transition. We developed a translational approach of AKI-to-CKD transition by reproducing human AAN in rodent models. Indeed, in such models, an early phase of acute tubular necrosis was rapidly followed by a massive interstitial recruitment of activated monocytes/macrophages followed by cytotoxic T lymphocytes, resulting in a transient AKI episode. A later chronic phase was then observed with progressive tubular atrophy related to dedifferentiation and necrosis of tubular epithelial cells. The accumulation of vimentin and αSMA-positive cells expressing TGFß in interstitial areas suggested an increase in resident fibroblasts and their activation into myofibroblasts resulting in collagen deposition and CKD. In addition, we identified 4 major actors in the AKI-to-CKD transition: (1) the tubular epithelial cells, (2) the endothelial cells of the interstitial capillary network, (3) the inflammatory infiltrate, and (4) the myofibroblasts. This review provides the most comprehensive and informative data we were able to collect and examines the pending questions.

14.
Nephron Exp Nephrol ; 118(3): e60-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21228600

RESUMEN

AIMS: We used a rat model of renal ischemia (35 min) to test the potential involvement of platelet/endothelial cell adhesion molecule 1 (PECAM-1/CD31) in the process of S3 tubule regeneration. METHODS: A monoclonal antibody specific for murine PECAM-1 was injected i.p. immediately after kidney reperfusion or 48 h post-ischemia. One day before ischemia, each animal received an i.p. injection of 80 mg/kg 5-bromo-2'-deoxyuridine (BrdU). Experimental animals were sacrificed 1, 2, 3, 7 and 14 days post-ischemia. Renal sections were processed to characterize the histopathological alterations and the distribution of BrdU-immunopositive cells. RESULTS: Our observations showed that anti-PECAM-1 administration was associated with an inhibition of S3 tubule regeneration along with a progressive cystic dilatation of renal tubules that was particularly prominent 2 weeks post-ischemia. Interestingly, injection of anti-PECAM-1 48 h post-ischemia failed to block renal regeneration and was followed by a normal re-epithelialization of S3 tubules. CONCLUSION: Our data showed that the blockade of PECAM-1 immediately after kidney reperfusion inhibits tubular regeneration. These observations suggest that transendothelial migration of extrarenal cells could be a precocious and pivotal step in kidney reparation, but also suggest that these extrarenal cells could be essential to the process of tubular regeneration.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Túbulos Renales/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Daño por Reperfusión/inmunología , Animales , Necrosis Tubular Aguda/inmunología , Necrosis Tubular Aguda/patología , Masculino , Ratones , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Ratas , Ratas Wistar , Regeneración/efectos de los fármacos , Regeneración/inmunología , Daño por Reperfusión/tratamiento farmacológico , Migración Transendotelial y Transepitelial
15.
Colloids Surf B Biointerfaces ; 203: 111770, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33894650

RESUMEN

Designing new materials to encapsulate living therapeutic cells for the treatment of the diseases caused by protein or hormone deficiencies is a great challenge. The desired materials need to be biocompatible towards both entrapped cells and host organisms, have long-term in vivo stability after implantation, allow the diffusion of nutrients and metabolites, and ensure perfect immune-isolation. The current work investigates the in vivo biocompatibility and stability of alginate@TiO2 hybrid microcapsules and the immune-isolation of entrapped HepG2 cells, to assess their potential for cell therapy. A comparison was made with alginate-silica hybrid microcapsules (ASA). These two hybrid microcapsules are implanted subcutaneously in female Wistar rats. The inflammatory responses of the rats are monitored by the histological examination of the implants and the surrounding tissues, to indicate their in vivo biocompatibility towards the hosts. The in vivo stability of the microcapsules is evaluated by the recovery rate of the intact microcapsules after implantation. The immune-isolation of the entrapped cells is assessed by their morphology, membrane integrity and intracellular enzymatic activity. The results show high viability of the entrapped cells and insignificant inflammation of the hosts, suggesting the excellent biocompatibility of alginate@TiO2 and ASA microcapsules towards both host organisms and entrapped cells. Compared to the ASA microcapsules, more intact alginate@TiO2 hybrid microcapsules are recovered 2-day and 2-month post-implantation and more cells remain alive, proving their better in vivo biocompability, stability, and immune-isolation. The present study demonstrates that the alginate@TiO2 hybrid microcapsule is a highly promising implantation material for cell therapy.


Asunto(s)
Alginatos , Tratamiento Basado en Trasplante de Células y Tejidos , Animales , Materiales Biocompatibles , Cápsulas , Femenino , Ácido Glucurónico , Ácidos Hexurónicos , Ratas , Ratas Wistar , Titanio
16.
Nat Commun ; 12(1): 4829, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376643

RESUMEN

Plasma hyaluronan (HA) increases systemically in type 2 diabetes (T2D) and the HA synthesis inhibitor, 4-Methylumbelliferone, has been proposed to treat the disease. However, HA is also implicated in normal physiology. Therefore, we generated a Hyaluronan Synthase 2 transgenic mouse line, driven by a tet-response element promoter to understand the role of HA in systemic metabolism. To our surprise, adipocyte-specific overproduction of HA leads to smaller adipocytes and protects mice from high-fat-high-sucrose-diet-induced obesity and glucose intolerance. Adipocytes also have more free glycerol that can be released upon beta3 adrenergic stimulation. Improvements in glucose tolerance were not linked to increased plasma HA. Instead, an HA-driven systemic substrate redistribution and adipose tissue-liver crosstalk contributes to the systemic glucose improvements. In summary, we demonstrate an unexpected improvement in glucose metabolism as a consequence of HA overproduction in adipose tissue, which argues against the use of systemic HA synthesis inhibitors to treat obesity and T2D.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Dioxoles/farmacología , Glucosa/metabolismo , Ácido Hialurónico/metabolismo , Lipólisis/efectos de los fármacos , Adipocitos/citología , Tejido Adiposo/citología , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Femenino , Intolerancia a la Glucosa/metabolismo , Homeostasis , Humanos , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo
17.
J Biomed Biotechnol ; 2010: 193259, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20617137

RESUMEN

This work explores the distribution of various markers expressed by interstitial cells in rat kidneys after ischemic injury (35 minutes) during regeneration of S3 tubules of outer stripe of outer medulla (OSOM). Groups of experimental animals (n = 4) were sacrificed every two hours during the first 24 hours post-ischemia as well as 2, 3, 7, 14 days post-ischemia. The occurrence of lineage markers was analyzed on kidney sections by immunohistochemistry and morphometry during the process of tubular regeneration. In postischemic kidneys, interstitial cell proliferation, assessed by 5-bromo-2'-deoxyuridine (BrdU) and Proliferating Cell Nuclear Antigen (PCNA) labeling, was prominent in outer medulla and reach a maximum between 24 and 72 hours after reperfusion. This population was characterized by the coexpression of vimentin and nestin. The density of -Neural Cell Adhesion Molecule (NCAM) positive interstitial cells increased transiently (18-72 hours) in the vicinity of altered tubules. We have also localized a small population of alpha-Smooth Muscle Actin (SMA)-positive cells confined to chronically altered areas and characterized by a small proliferative index. In conclusion, we observed in the postischemic kidney a marked proliferation of interstitial cells that underwent transient phenotypical modifications. These interstitial cells could be implicated in processes leading to renal fibrosis.


Asunto(s)
Proteínas de Filamentos Intermediarios/metabolismo , Isquemia/metabolismo , Túbulos Renales Proximales , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Vimentina/metabolismo , Actinas/metabolismo , Animales , Bromodesoxiuridina , Proliferación Celular , Inmunohistoquímica , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Masculino , Microscopía Fluorescente , Nestina , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Wistar , Fase S
18.
Nephrol Dial Transplant ; 23(12): 3786-97, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18678561

RESUMEN

BACKGROUND: In this study, we have examined rat kidneys after ischaemic injury (35 min) with regard to the dynamics of S3 tubule regeneration. METHODS: One day before ischaemia, each rat received four successive i.p. injections of BrdU (5-bromo-2'-deoxyuridine: 80 mg/kg) at 2 h intervals. Groups of experimental animals (n = 4) were killed every 2 h during the first 24 h post-ischaemia as well as 2, 3, 7 and 14 days post-ischaemia. Renal sections were processed to characterize by immunohistochemistry the distribution and phenotype of BrdU-positive cells. RESULTS: Renal regeneration after ischaemia was associated with a typical sequence of transient events: (1) absence of immunostaining during the first 8 h after reperfusion; (2) between 8 and 16 h, detection of a small population of BrdU-positive cells (CD44(+), vimentin(+), CD45(-)) restricted to the lumen of blood vessels characterized by the endothelial expression of selectin E; (3) between 16 and 24 h, progressive decrease of labelled cells in renal capillaries and a concomitant increase in the interstitial compartment; (4) after 1 day, labelled cells disappeared progressively from peritubular interstitium and were mainly observed in regenerating S3 tubules, and (5) after 3 days numerous positive cells were only present in regenerated tubules. CONCLUSIONS: Our data suggest that positive cells (BrdU(+), CD44(+), vimentin(+) and CD45(-)) observed in kidney tubules after ischaemia could originate from an extrarenal source and reach the renal parenchyma via blood vessels. We postulate that these immature cells migrate to injured tubules, proliferate and finally differentiate into mature epithelial cells leading to the replacement of a majority (>80%) of altered S3 cells.


Asunto(s)
Riñón/irrigación sanguínea , Riñón/lesiones , Actinas/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Bromodesoxiuridina/metabolismo , Movimiento Celular , Selectina E/metabolismo , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Inmunohistoquímica , Riñón/fisiopatología , Necrosis Tubular Aguda/etiología , Necrosis Tubular Aguda/patología , Necrosis Tubular Aguda/fisiopatología , Túbulos Renales/patología , Túbulos Renales/fisiopatología , Antígenos Comunes de Leucocito/metabolismo , Masculino , Cadenas Pesadas de Miosina/metabolismo , Ratas , Ratas Wistar , Regeneración , Daño por Reperfusión/complicaciones , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Vimentina/metabolismo
19.
PLoS One ; 12(8): e0183604, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28832640

RESUMEN

Aristolochic Acid (AA) nephropathy (AAN) is a progressive tubulointerstitial nephritis characterized by an early phase of acute kidney injury (AKI) leading to chronic kidney disease (CKD). The reduced nitric oxide (NO) bioavailability reported in AAN might contribute to renal function impairment and progression of the disease. We previously demonstrated that L-arginine (L-Arg) supplementation is protective in AA-induced AKI. Since the severity of AKI may be considered a strong predictor of progression to CKD, the present study aims to assess the potential benefit of L-Arg supplementation during the transition from the acute phase to the chronic phase of AAN. C57BL/6J male mice were randomly subjected to daily i.p. injections of vehicle or AA for 4 days. To determine whether renal AA-induced injuries were linked to reduced NO production, L-Arg was added to drinking water from 7 days before starting i.p. injections, until the end of the protocol. Mice were euthanized 5, 10 and 20 days after vehicle or AA administration. AA-treated mice displayed marked renal injury and reduced NO bioavailability, while histopathological features of AAN were reproduced, including interstitial cell infiltration and tubulointerstitial fibrosis. L-Arg treatment restored renal NO bioavailability and reduced the severity of AA-induced injury, inflammation and fibrosis. We concluded that reduced renal NO bioavailability contributes to the processes underlying AAN. Furthermore, L-Arg shows nephroprotective effects by decreasing the severity of acute-to-chronic transition in experimental AAN and might represent a potential therapeutic tool in the future.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Enfermedades Renales/metabolismo , Óxido Nítrico/metabolismo , Animales , Arginina/administración & dosificación , Disponibilidad Biológica , Inflamación/prevención & control , Enfermedades Renales/inducido químicamente , Ratones
20.
Int J Mol Med ; 18(1): 83-94, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16786159

RESUMEN

Ischemia/reperfusion (I/R) injury in the kidney involves hemodynamic and cellular dysfunctions as well as leukocyte infiltration. Functional recovery occurs via cell proliferation and/or migration. To determine the roles of hyaluronan (HA) and its main receptor CD44 in renal postischemic processes, we compared their localization and expression with that of neutrophils, macrophages, and PCNA-positive (regenerative) cells as characterized by immunohistochemistry, up to 28 days after I/R in uninephrectomized rats. Observations covered all kidney zones, i.e. cortex (C), outer and inner stripes of outer medulla (OSOM, ISOM), and inner medulla (IM). In controls, HA was localized to the interstitium of IM and ISOM, and CD44 was mostly present on the basolateral membranes of collecting ducts in ISOM, the thin descending limb of Henle's loop and macula densa cells. After I/R, HA and CD44 staining appeared in C and OSOM at 12 h and persisted throughout the regenerative period, i.e. until day 7. Thereafter, they regressed but remained associated with remodeling areas. CD44 expression was found de novo on the apical pole of regenerating, not fully differentiated tubular cells and on some interstitial cells. It was prominent on all infiltrating neutrophils, as soon as 2 h post-I/R, and on 30% of the macrophages, including those in late HA-rich inflammatory granulomas. CD44 is probably involved in early leukocyte infiltration, in tubular regeneration, and in macrophage activity, while HA modifies the physico-chemical environment of interstitial and migrating cells. Based on its presence in remodeling areas, the HA-CD44 pair may be implicated in persistent postichemic inflammation as observed in chronic allograft nephropathy.


Asunto(s)
Receptores de Hialuranos/análisis , Ácido Hialurónico/análisis , Daño por Reperfusión/metabolismo , Animales , Acuaporina 3/análisis , Modelos Animales de Enfermedad , Ectodisplasinas , Inmunohistoquímica , Riñón/irrigación sanguínea , Riñón/química , Riñón/patología , Corteza Renal/química , Corteza Renal/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Médula Renal/química , Médula Renal/patología , Túbulos Renales/química , Túbulos Renales/patología , Masculino , Proteínas de la Membrana/análisis , Neutrófilos/química , Neutrófilos/patología , Ratas , Ratas Wistar , Daño por Reperfusión/patología , Factores de Tiempo , Factores de Necrosis Tumoral/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA