Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39216659

RESUMEN

Demonstrating drug efficacy in slowing kidney disease progression requires large clinical trials when targeting participants with an early stage of chronic kidney disease (CKD). In this randomized, parallel-group, open-labeled trial (CANPIONE study), we assessed the effect of the sodium-glucose cotransporter 2 (SGLT2) inhibitor canagliflozin using the individual's change in estimated glomerular filtration rate (eGFR) slope before (pre-intervention slope) and during treatment (chronic slope). We randomly assigned (1:1) participants with type 2 diabetes, urinary albumin-to-creatinine ratio (UACR) of 50 to under 300 mg/g, and an eGFR of at least 45 ml/min/1.73m2 to receive canagliflozin or guideline-recommended treatment except for SGLT2 inhibitors (control). The first and second primary outcomes were the geometric mean percentage change from baseline in UACR and the change in eGFR slope, respectively. Of 98 randomized participants, 96 received at least one study treatment. The least-squares mean change from baseline in log-transformed geometric mean UACR was significantly greater in the canagliflozin group than the control group (between group-difference, -30.8% (95% confidence interval -42.6 to -16.8). The between-group difference (canagliflozin group - control group) of change in eGFR slope (chronic - pre-intervention) was 4.4 (1.6 to 7.3) ml/min/1.73 m2 per year, which was more pronounced in participants with faster eGFR decline. In summary, canagliflozin reduced albuminuria and the participant-specific natural course of eGFR decline in participants with type 2 diabetes and microalbuminuria. Thus, the CANPIONE study suggests that the within-individual change in eGFR slope may be a novel approach to determine the kidney protective potential of new therapies in early stages of CKD.

2.
Diabetes Obes Metab ; 24(8): 1429-1438, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35491532

RESUMEN

AIM: To evaluate the effect of canagliflozin, a sodium-glucose co-transporter-2 (SGLT2) inhibitor, on albuminuria and the decline of estimated glomerular filtration rate (eGFR) in participants with type 2 diabetes and microalbuminuria. METHODS: The CANPIONE study is a multicentre, randomized, parallel-group and open-labelled study consisting of a unique 24-week preintervention period, during which the rate of eGFR decline before intervention is estimated, followed by a 52-week intervention and a 4-week washout period. Participants with a geometric mean urinary albumin-to-creatinine ratio (UACR) of 50 and higher and less than 300 mg/g in two consecutive first-morning voids at two different time points, and an eGFR of 45 ml/min/1.73m2 or higher, are randomly assigned to receive canagliflozin 100 mg daily or to continue guideline-recommended treatment, except for SGLT2 inhibitors. The first primary outcome is the change in UACR, and the second primary outcome is the change in eGFR slope. RESULTS: A total of 258 participants were screened and 98 were randomized at 21 sites in Japan from August 2018 to May 2021. The mean baseline age was 61.4 years and 25.8% were female. The mean HbA1c was 7.9%, mean eGFR was 74.1 ml/min/1.73m2 and median UACR was 104.2 mg/g. CONCLUSIONS: The CANPIONE study will determine whether the SGLT2 inhibitor canagliflozin can reduce albuminuria and slow eGFR decline in participants with type 2 diabetes and microalbuminuria.


Asunto(s)
Diabetes Mellitus Tipo 2 , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Albuminuria/epidemiología , Canagliflozina/uso terapéutico , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/orina , Femenino , Tasa de Filtración Glomerular , Humanos , Japón/epidemiología , Masculino , Persona de Mediana Edad , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
3.
Angiogenesis ; 23(4): 667-684, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32699964

RESUMEN

Platelet-derived growth factor-B (PDGF-B) is a main factor to promote adipose tissue angiogenesis, which is responsible for the tissue expansion in obesity. In this process, PDGF-B induces the dissociation of pericytes from blood vessels; however, its regulatory mechanism remains unclear. In the present study, we found that stromal cell-derived factor 1 (SDF1) plays an essential role in this regulatory mechanism. SDF1 mRNA was increased in epididymal white adipose tissue (eWAT) of obese mice. Ex vivo pharmacological analyses using cultured adipose tissue demonstrated that physiological concentrations (1-100 pg/mL) of SDF1 inhibited the PDGF-B-induced pericyte dissociation from vessels via two cognate SDF1 receptors, CXCR4 and CXCR7. In contrast, higher concentrations (> 1 ng/mL) of SDF1 alone caused the dissociation of pericytes via CXCR4, and this effect disappeared in the cultured tissues from PDGF receptor ß (PDGFRß) knockout mice. To investigate the role of SDF1 in angiogenesis in vivo, the effects of anagliptin, an inhibitor of dipeptidyl peptidase 4 (DPP4) that degrades SDF1, were examined in mice fed a high-fat diet. Anagliptin increased the SDF1 levels in the serum and eWAT. These changes were associated with a reduction of pericyte dissociation and fat accumulation in eWAT. AMD3100, a CXCR4 antagonist, cancelled these anagliptin effects. In flow-cytometry analysis, anagliptin increased and decreased the PDGF-B expression in endothelial cells and macrophages, respectively, whereas anagliptin reduced the PDGFRß expression in pericytes of eWAT. These results suggest that SDF1 negatively regulates the adipose tissue angiogenesis in obesity by altering the reactivity of pericytes to PDGF-B.


Asunto(s)
Tejido Adiposo Blanco/patología , Tejido Adiposo Blanco/fisiopatología , Quimiocina CXCL12/metabolismo , Obesidad/patología , Obesidad/fisiopatología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Remodelación Vascular , Tejido Adiposo Blanco/irrigación sanguínea , Inductores de la Angiogénesis/metabolismo , Animales , Vasos Sanguíneos/patología , Quimiocina CXCL12/sangre , Dieta Alta en Grasa , Epidídimo/patología , Conducta Alimentaria , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Modelos Biológicos , Neovascularización Fisiológica/efectos de los fármacos , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Pericitos/patología , Pirimidinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Delgadez/patología
4.
Int J Mol Sci ; 21(3)2020 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-31991851

RESUMEN

Dipeptidyl peptidase (DPP)-4, a molecular target of DPP-4 inhibitors, which are type 2 diabetes drugs, is expressed in a variety of cell types, tissues and organs. DPP-4 has been shown to be involved in cancer biology, and we have recently shown that a DPP-4 inhibitor promoted the epithelial mesenchymal transition (EMT) in breast cancer cells. The EMT is known to associate with chemotherapy resistance via the induction of ATP-binding cassette (ABC) transporters in cancer cells. Here, we demonstrated that deficiency in DPP-4 promoted chemotherapy resistance via the CXCL12/CXCR4/mTOR axis, activating the TGFß signaling pathway via the expression of ABC transporters. DPP-4 inhibition enhanced ABC transporters in vivo and in vitro. Doxorubicin (DOX) further induced ABC transporters in DPP-4-deficient 4T1 cells, and the induction of ABC transporters was suppressed by either the CXCR4 inhibitor AMD3100, the mTOR inhibitor rapamycin or a neutralizing TGFß (1, 2 and 3) antibody(N-TGFß). Knockdown of snail, an EMT-inducible transcription factor, suppressed ABC transporter levels in DOX-treated DPP-4-deficient 4T1 cells. In an allograft mouse model, however, the effects of DOX in either primary tumor or metastasis were not statistically different between control and DPP-4-kd 4T1. Taken together, our findings suggest that DPP-4 inhibitors potentiate chemotherapy resistance via the induction of ABC transporters by the CXCL12/CXCR4/mTOR/TGFß signaling pathway in breast cancer cells.


Asunto(s)
Quimiocina CXCL12/metabolismo , Dipeptidil Peptidasa 4/deficiencia , Resistencia a Antineoplásicos , Proteínas de Neoplasias , Receptores CXCR4/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Quimiocina CXCL12/genética , Dipeptidil Peptidasa 4/metabolismo , Femenino , Humanos , Células MCF-7 , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores CXCR4/genética , Serina-Treonina Quinasas TOR/genética , Factor de Crecimiento Transformador beta/genética
5.
Clin Exp Nephrol ; 23(8): 1004-1012, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30949886

RESUMEN

BACKGROUND: A biomarker, by which we can predict alterations of renal function in normoalbuminuric diabetic patients, is not available. Here, we report that endogenous anti-fibrotic peptide N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) represents a potential biomarker to predict alterations in eGFR in normoalbuminuric diabetic patients. METHODS: We analyzed 21 normoalbuminuric diabetic patients with eGFR ≥ 30 ml/min/1.73 m2 and measured AcSDKP levels in first morning void urine. We divided patients into two groups based on the median values: low or high urinary AcSDKP groups (uAcSDKP/Crlow or uAcSDKP/Crhigh). At baseline, no significant differences in sex, age, HbA1c, BMI, serum creatinine levels, etc., were observed between the two groups. RESULTS: During ~ 4 years, the alteration in eGFR [ΔeGFRop (ΔeGFR observational periods)] was significantly stable in uAcSDKP/Crhigh group compared with uAcSDKP/Crlow group over time (P = 0.003, χ2 = 8.58). We also evaluated urine kidney injury molecule-1 (uKim-1) levels and found that ΔeGFRop was also stable in low uKim-1 group compared with high uKim-1 group over time (P = 0.004, χ2 = 8.38). Patients who fulfilled the criteria for both uAcSDKP/Crhigh and uKim-1low exhibited stable ΔeGFRop (P < 0.001, χ2 = 30.4) when compared to the remaining patients. Plasma AcSDKP (P = 0.015, χ2 = 5.94) and urine ß2-microglobulin (P = 0.038, χ2 = 4.31) also display weak but significant predictor of ΔeGFRop as well. CONCLUSION: AcSDKP represents a potentially useful biomarker to predict alterations in the renal function of patients with diabetes presenting normoalbuminuria.


Asunto(s)
Diabetes Mellitus Tipo 2/orina , Nefropatías Diabéticas/orina , Tasa de Filtración Glomerular , Riñón/fisiopatología , Oligopéptidos/orina , Anciano , Biomarcadores/orina , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/fisiopatología , Nefropatías Diabéticas/diagnóstico , Nefropatías Diabéticas/fisiopatología , Diagnóstico Precoz , Femenino , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Factores de Tiempo , Urinálisis
6.
Biochem Biophys Res Commun ; 495(3): 2214-2220, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29269295

RESUMEN

Fibroblast growth factor receptor (FGFR) 1 plays a key role in endothelial homeostasis by inducing microRNA (miR) let-7. Our previous paper showed that anti-fibrotic effects of N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) were associated with restoring diabetes-suppressed expression of FGFR1 and miR let-7, the key contributor of mitochondrial biogenesis, which is regulated by mitochondrial membrane GTPase proteins (MFN2 and OPA1). Here, we found that the FGFR1 signaling pathway was critical for AcSDKP in maintaining endothelial mitochondrial biogenesis through induction of miR let-7b-5p. In endothelial cells, AcSDKP restored the triple cytokines (TGF-ß2, interleukin-1ß, tumor necrosis factor-α)-suppressed miR let-7b-5p and protein levels of the mitochondrial membrane GTPase. This effect of AcSDKP was lost with either fibroblast growth factor receptor substrate 2 (FRS2) siRNA or neutralizing FGFR1-treated cells. Similarly, AcSDKP had no effect on the miR let-7b-5p inhibitor-suppressed GTPase levels in endothelial cells. In addition, a miR let-7b-5p mimic restored the levels of FRS2 siRNA-reduced GTPases in endothelial cells. These findings were also confirmed using MitoTracker Green and an immunofluorescence assay. Our results demonstrated that the AcSDKP-FGFR1 signaling pathway is critical for maintaining mitochondrial dynamics by control of miR let-7b-5p in endothelial cells.


Asunto(s)
Células Endoteliales/fisiología , MicroARNs/metabolismo , Mitocondrias/fisiología , Oligopéptidos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Humanos , Regulación hacia Arriba/fisiología
7.
Biochim Biophys Acta Gen Subj ; 1862(7): 1547-1555, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29550428

RESUMEN

Muscle biology is important topic in diabetes research. We have reported that a diet with ketogenic amino acids rich replacement (KAAR) ameliorated high-fat diet (HFD)-induced hepatosteatosis via activation of the autophagy system. Here, we found that a KAAR ameliorated the mitochondrial morphological alterations and associated mitochondrial dysfunction induced by an HFD through induction of the AKT/4EBP1 and autophagy signaling pathways in both fast and slow muscles. The mice were fed with a standard HFD (30% fat in food) or an HFD with KAAR (HFDKAAR). In both the gastrocnemius and the soleus, HFDKAAR ameliorated HFD-impaired mitochondrial morphology and mitochondrial function, characterized by decreased mitofusin 2, optic atrophy 1, peroxisome proliferator-activated receptor (PPAR) γ coactivator-1α and PPARα levels and increased dynamin-related protein 1 levels. The decreased levels of phosphorylated AKT and 4EBP1 in the gastrocnemius and soleus of HFD-fed mice were remediated by HFDKAAR. Furthermore, the HFDKAAR ameliorated the HFD-induced autophagy defects in the gastrocnemius and soleus. These findings suggest that KAAR may be a novel strategy to combat obesity-induced mitochondrial dysfunction, likely through induction of the AKT/4EBP1 and autophagy pathways in skeletal muscle.


Asunto(s)
Aminoácidos/farmacología , Autofagia/fisiología , Proteínas Portadoras/fisiología , Dieta Cetogénica , Mitocondrias/fisiología , Músculo Esquelético/fisiología , Obesidad/dietoterapia , Fosfoproteínas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Aminoácidos/administración & dosificación , Animales , Autofagia/efectos de los fármacos , Sangre , Glucemia/análisis , Peso Corporal/efectos de los fármacos , Proteínas de Ciclo Celular , Factores Eucarióticos de Iniciación , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Obesidad/fisiopatología , Tamaño de los Órganos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
8.
Curr Diab Rep ; 17(7): 53, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28593583

RESUMEN

PURPOSE OF REVIEW: Autophagy promotes cellular health in response to various cellular stresses and to changes in nutrient conditions. In this review, we focus on the role of autophagy in the pathogenesis of diabetic nephropathy and discuss the regulation of autophagy as a new therapeutic target for the suppression of diabetic nephropathy. RECENT FINDINGS: Previous studies have indicated that autophagy deficiency or insufficiency in renal cells, including podocytes, mesangial cells, endothelial cells and tubular cells, contributes to the pathogenesis of diabetic nephropathy. Alterations in the nutrient-sensing pathways, including mammalian target of rapamycin complex1 (mTORC1), AMP-activated kinase (AMPK) and Sirt1, due to excess nutrition in diabetes are implicated in the impairment of autophagy. Maintaining both basal and adaptive autophagy against cellular stress may protect the kidney from diabetes-induced cellular stresses. Therefore, the activation of autophagy through the modulation of nutrient-sensing pathways may be a new therapeutic option for the suppression of diabetic nephropathy.


Asunto(s)
Autofagia , Nefropatías Diabéticas/patología , Envejecimiento/patología , Animales , Humanos , Células Mesangiales/patología , Modelos Biológicos , Podocitos/patología
9.
Nephrology (Carlton) ; 22(12): 1030-1034, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28802091

RESUMEN

A low-protein diet (LPD), particularly, very low-protein diet (VLPD) is expected for reno-protection in advanced chronic kidney disease, including diabetic nephropathy. We previously also demonstrated that a VLPD clearly improved advanced diabetic nephropathy in a type 2 diabetes and obesity rat. However, clinically, an everyday long-term VLPD contributes to poor adherence, which may be related to controvertial results of an LPD on the suppression for diabetic nephropathy, and has nutritional issues, such as sarcopenia or protein-energy wasting. The aim of this study is to elucidate the reno-protective effect of a cyclic and intermittent VLPD, not an everyday VLPD, against the advanced experimental diabetic nephropathy. Diabetic male Wistar fatty (fa/fa) rats (WFRs) were treated with a standard diet (STD; 23.84% protein) or a cyclic and intermittent VLPD (5.77% protein) consisting of an STD for 3 days and a VLPD for 4 days a week for 20 weeks beginning at 24 weeks of age. A cyclic and intermittent VLPD significantly improved renal hypertrophy, and significantly decreased urinary albumin and liver-type fatty acid binding protein (L-FABP) excretion without changes in body weight or exacerbation of HbA1c levels in diabetic rats. Additionally, diabetes-induced renal injuries including fibrosis, tubular cell damage and inflammation were significantly ameliorated by a cyclic and intermittent VLPD in diabetic rats. Thus, based on our experimental data, a cyclic and intermittent VLPD may be a dietary regimen that is easy to continue and has less risk of malnutrition, compared to an everyday long-term VLPD, against advanced diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/dietoterapia , Proteínas en la Dieta/administración & dosificación , Obesidad/complicaciones , Animales , Modelos Animales de Enfermedad , Femenino , Hemoglobina Glucada/análisis , Masculino , Ratas , Ratas Wistar
10.
Diabetologia ; 59(6): 1307-17, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27020449

RESUMEN

AIMS/HYPOTHESIS: The efficacy of a low-protein diet (LPD) on diabetic nephropathy is controversial. We aimed to investigate the renoprotective effects of an LPD and the underlying molecular mechanism in a rat model of type 2 diabetes and obesity. METHODS: Diabetic male Wistar fatty (fa/fa) rats (WFRs) were treated with a standard diet (23.84% protein) or an LPD (5.77% protein) for 20 weeks from 24 weeks of age. We investigated the effect of the LPD on renal function, fibrosis, tubular cell damage, inflammation, mitochondrial morphology of proximal tubular cells (PTCs), apoptosis, autophagy and activation of mammalian target of rapamycin complex 1 (mTORC1). RESULTS: Kidney weight, albuminuria, excretion of urinary liver-type fatty acid binding protein, levels of plasma cystatin C and changes in renal histology, including fibrosis, tubular cell damage and inflammation, were aggravated in WFRs compared with non-diabetic Wistar lean rats (WLRs). Fragmented and swelling mitochondria accumulated in PTCs and apoptosis were enhanced in the kidney of WFRs. Immunohistochemical staining of p62 and p-S6 ribosomal protein (p-S6RP) in the tubular lesions of WFRs was increased compared with WLRs. The LPD intervention clearly ameliorated damage as shown by the assessment of renal function and histology, particularly tubulointerstitial damage in diabetic kidneys. Additionally, the 5.77% LPD, but not the 11.46% LPD, significantly suppressed p-S6RP levels and increased microtubule-associated protein light chain 3-II levels in the renal cortex. The LPD intervention partially decreased HbA1c levels in WFRs, and no differences in mean BP were observed among any of the groups. CONCLUSIONS/INTERPRETATION: A very-low-protein diet improved advanced diabetic renal injuries, including tubulointerstitial damage, by restoring autophagy through the suppression of the mTORC1 pathway.


Asunto(s)
Autofagia/fisiología , Diabetes Mellitus Tipo 2/dietoterapia , Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/dietoterapia , Nefropatías Diabéticas/metabolismo , Dieta con Restricción de Proteínas , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/genética , Diabetes Mellitus Tipo 2/patología , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Microscopía Electrónica de Transmisión , Complejos Multiproteicos/genética , Obesidad/dietoterapia , Obesidad/metabolismo , Obesidad/patología , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Serina-Treonina Quinasas TOR/genética
11.
Kidney Int ; 88(3): 479-89, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25830763

RESUMEN

Integrin ß1 and dipeptidyl peptidase (DPP)-4 play roles in endothelial cell biology. Vascular endothelial growth factor (VEGF)-A inhibits endothelial-to-mesenchymal transition (EndMT) through VEGF-R2, but through VEGF-R1 promotes EndMT by reducing the bioavailability of VEGF-A. Here we tested whether DPP-4-integrin ß1 interactions have a role in EndMT in the renal fibrosis of diabetic nephropathy. In streptozotocin-induced fibrotic kidneys in diabetic CD-1 mice, levels of endothelial DPP-4, integrin ß1, and phospho-integrin ß1 were all higher and associated with plasma cystatin C elevation. The DPP-4 inhibitor linagliptin ameliorated kidney fibrosis, reduced plasma cystatin C levels, and suppressed endothelial levels of DPP-4, integrin ß1, and phospho-integrin ß1. In cultured endothelial cells, DPP-4 and integrin ß1 physically interacted. Suppression of DPP-4 by siRNA was associated with suppression of integrin ß1 and vice versa. Knockdown of either integrin ß1 or DPP-4 resulted in the silencing of TGF-ß2-induced TGF-ß receptor heterodimer formation, smad3 phosphorylation, and EndMT. DPP-4 negatively regulated endothelial viability signaling by VEGF-R2 suppression and VEGF-R1 induction in endothelial cells. Thus, DPP-4 and integrin ß1 interactions regulate key endothelial cell signal transduction in both physiological and pathological conditions including EndMT. Hence, inhibiting DPP-4 may be a therapeutic target for treating kidney fibrosis in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Nefropatías Diabéticas/enzimología , Dipeptidil Peptidasa 4/metabolismo , Células Endoteliales/enzimología , Transición Epitelial-Mesenquimal , Integrina beta1/metabolismo , Riñón/enzimología , Animales , Supervivencia Celular , Células Cultivadas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/prevención & control , Dipeptidil Peptidasa 4/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibrosis , Hipoglucemiantes/farmacología , Integrina beta1/genética , Riñón/efectos de los fármacos , Riñón/patología , Linagliptina/farmacología , Masculino , Ratones , Fosforilación , Interferencia de ARN , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Transfección
12.
Nihon Rinsho ; 73(12): 2037-43, 2015 Dec.
Artículo en Japonés | MEDLINE | ID: mdl-26666150

RESUMEN

Diabetic nephropathy(DN) is the most common cause of chronic kidney disease, leading to end-stage renal disease (ESRD) and cardiovascular disease (CVD). The overall number of patients with DN will continue to increase in parallel with the increasing global pandemic of type 2 diabetes. The detection of albuminuria is most important for diagnosis of early stage of DN, and also estimated glomerular filtration rate (eGFR) should be assessed, because both albuminuria and reduction of GFR are recognized as the independent risk factor for progression of ESRD and initiation of CVD, respectively. Based on landmark clinical trials, both DN and CVD have become preventable by controlling conventional factors, including hyperglycemia targeting HbA1c<7.0%, hypertenstion using renin angiotensin system inhibitors, dyslipidemia using statins or fibrates, and multifactorial treatment.


Asunto(s)
Nefropatías Diabéticas/terapia , Albuminuria/diagnóstico , Enfermedades Cardiovasculares/etiología , Nefropatías Diabéticas/diagnóstico , Tasa de Filtración Glomerular , Humanos , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Metabolismo de los Lípidos , Insuficiencia Renal Crónica/etiología
13.
Biochim Biophys Acta ; 1830(10): 4820-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23800577

RESUMEN

BACKGROUND: Calorie restriction (CR) is accepted as an experimental anti-aging paradigm. Several important signal transduction pathways including AMPK and SIRT1 are implicated in the regulation of physiological processes of CR. However, the mechanisms responsible for adaptations remain unclear in humans. SCOPE OF REVIEW: Four overweight male participants were enrolled and treated with 25% CR of their baseline energy requirements for 7weeks. Characteristics, including body weight (BW), body mass index (BMI), %fat, visceral fat area (VFA), mean blood pressure (MBP) and VO2 max, as well as metabolic parameters, such as insulin, lipid profiles and inflammatory makers and the expression of phosphorylated AMPK and SIRT1 in peripheral blood mononuclear cells (PBMNCs), were determined at baseline and then after 7weeks. In addition, we assessed the effects of the serum collected from the participants on AMPK and SIRT1 activation and mitochondrial biogenesis in cultured human skeletal muscle cells. MAJOR CONCLUSIONS: After CR, BW, BMI, %fat, VFA and MBP all significantly decreased, while VO2 max increased, compared to those at baseline. The levels of fasting insulin, free fatty acid, and inflammatory makers, such as interleukin-6 and visfatin, were significantly reduced, whereas the expression of phosphorylated AMPK and SIRT1 was significantly increased in PBMNCs collected after CR, compared to those at baseline. The skeletal muscle cells that were cultured in serum collected after CR showed an increase in AMPK and SIRT1 activity as well as mitochondrial biogenesis. GENERAL SIGNIFICANCE: CR is beneficial for obesity-related metabolic alterations and induces cellular adaptations against aging, possibly through AMPK and SIRT1 activation via circulating factors.


Asunto(s)
Adenilato Quinasa/metabolismo , Restricción Calórica , Enfermedades Metabólicas/complicaciones , Obesidad/complicaciones , Sobrepeso/dietoterapia , Sirtuina 1/metabolismo , Adulto , Células Cultivadas , Activación Enzimática , Humanos , Masculino , Enfermedades Metabólicas/enzimología , Enfermedades Metabólicas/metabolismo , Persona de Mediana Edad , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Obesidad/enzimología , Obesidad/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Biochim Biophys Acta ; 1832(10): 1605-12, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23669346

RESUMEN

Ketogenic amino acid (KAA) replacement diet has been shown to cure hepatic steatosis, a serious liver disease associated with diverse metabolic defects. In this study, we investigated the effects of KAA replacement diet on nutrition sensing signaling pathway and analyzed whether induction of hepatic autophagy was involved. Mice are fed with high fat diet (HFD) or KAA replacement in high-fat diet (30% fat in food; HFD)-fed (HFD(KAAR)) and sacrificed at 8, 12, 16 weeks after initiation of experimental food. Hepatic autophagy was analyzed in protein expression of several autophagy-associated molecules and in light chain-3 green fluorescent protein (LC-3 GFP) transgenic mice. HFD(KAAR) showed increased AMP-activated protein kinase (AMPK) phosphorylation and enhanced liver kinase B1 (LKB1) expression compared to control HFD-fed mice. The KAA-HFD-induced activation of AMPK was associated with an increased protein expression of sirtuin 1 (Sirt1), decreased forkhead box protein O3a (Foxo3a) level, and suppression of mammalian target of rapamycin (mTOR) phosphorylation compared with the HFD-fed mice. The intervention study revealed that a KAA-replacement diet also ameliorated all the established metabolic and autophagy defects in the HFD-fed mice, suggesting that a KAA-replacement diet can be used therapeutically in established diseases. These results indicate that KAA replacement in food could be a novel strategy to combat hepatic steatosis and metabolic abnormalities likely involvement of an induction of autophagy.


Asunto(s)
Aminoácidos Esenciales/administración & dosificación , Autofagia , Dieta Cetogénica , Hígado Graso/dietoterapia , Animales , Peso Corporal , Grasas de la Dieta/administración & dosificación , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos
15.
Curr Opin Nephrol Hypertens ; 23(1): 75-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24247822

RESUMEN

PURPOSE OF REVIEW: Accumulating evidence indicates the beneficial effects of sirtuins (SIRTs), including SIRT1 and SIRT3, which are NAD-dependent deacetylases, in age-related diseases such as diabetes, neuron disease, cardiovascular disease and kidney disease. RECENT FINDINGS: SIRT1 deacetylates many targets, such as transcriptional factors and proteins, and exhibits renoprotection through reduction of fibrosis, antiapoptotic and anti-inflammatory effects and induction of autophagy in renal cells. SIRT1 may also participate in the regulation of blood pressure by sodium handling and by decreasing the responsiveness for angiotensin II. However, SIRT1 may be involved in the progression of cyst formation of renal epithelial cells in autosomal-dominant polycystic kidney disease (ADPKD). SIRT3 protects renal tubular cells against palmitate-induced lipotoxicity through antioxidative and anti-inflammatory effects. SUMMARY: The activation of SIRT1 and SIRT3 may be a novel therapeutic strategy for kidney diseases, but not for ADPKD.


Asunto(s)
Enfermedades Renales/enzimología , Riñón/enzimología , Sirtuinas/metabolismo , Animales , Autofagia , Presión Sanguínea , Fibrosis , Humanos , Mediadores de Inflamación/metabolismo , Riñón/inmunología , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Enfermedades Renales/terapia , Transducción de Señal
16.
Clin Exp Nephrol ; 18(2): 210-3, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24221306

RESUMEN

Autophagy has evolved as a stress response that allows unicellular eukaryotic organisms to survive in starved conditions by regulating energy homeostasis and/or by protein and organelle quality control. The diabetes-induced accumulation of damaged proteins and organelles results in the development and progression of diabetic nephropathy. In contrast, autophagy machinery is activated by calorie restriction and environmental stress in proximal tubular cells, and is maintained at a high level in podocytes, suggesting its crucial role in the pathogenesis of diabetic nephropathy. However, its role in diabetic nephropathy has not been fully known. Here, we will discuss the role of autophagy and its involvement in the pathogenesis of diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/etiología , Adenilato Quinasa/metabolismo , Animales , Autofagia/fisiología , Nefropatías Diabéticas/tratamiento farmacológico , Podocitos/metabolismo , Sirtuinas/metabolismo
17.
Am J Physiol Endocrinol Metab ; 305(11): E1415-25, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24129399

RESUMEN

Because the renin-angiotensin-aldosterone system has been implicated in the development of insulin resistance and promotion of fibrosis in some tissues, such as the vasculature, we examined the effect of eplerenone, a selective mineralocorticoid receptor (MR) antagonist, on nonalcoholic steatohepatitis (NASH) and metabolic phenotypes in a mouse model reflecting metabolic syndrome in humans. We adopted liver-specific transgenic (Tg) mice overexpressing the active form of sterol response element binding protein-1c (SREBP-1c) fed a high-fat and fructose diet (HFFD) as the animal model in the present study. When wild-type (WT) C57BL/6 and liver-specific SREBP-1c Tg mice grew while being fed HFFD for 12 wk, body weight and epididymal fat weight increased in both groups with an elevation in blood pressure and dyslipidemia. Glucose intolerance and insulin resistance were also observed. Adipose tissue hypertrophy and macrophage infiltration with crown-like structure formation were also noted in mice fed HFFD. Interestingly, the changes noted in both genotypes fed HFFD were significantly ameliorated with eplerenone. HFFD-fed Tg mice exhibited the histological features of NASH in the liver, including macrovesicular steatosis and fibrosis, whereas HFFD-fed WT mice had hepatic steatosis without apparent fibrotic changes. Eplerenone effectively ameliorated these histological abnormalities. Moreover, the direct suppressive effects of eplerenone on lipopolysaccharide-induced TNFα production in the presence and absence of aldosterone were observed in primary-cultured Kupffer cells and bone marrow-derived macrophages. These results indicated that eplerenone prevented the development of NASH and metabolic abnormalities in mice by inhibiting inflammatory responses in both Kupffer cells and macrophages.


Asunto(s)
Hígado Graso/prevención & control , Síndrome Metabólico/tratamiento farmacológico , Antagonistas de Receptores de Mineralocorticoides/administración & dosificación , Espironolactona/análogos & derivados , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Animales , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Carbohidratos de la Dieta/administración & dosificación , Eplerenona , Hígado Graso/etiología , Fructosa/administración & dosificación , Hígado/metabolismo , Hígado/patología , Masculino , Síndrome Metabólico/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico , Fenotipo , Ratas , Ratas Wistar , Espironolactona/administración & dosificación
18.
Clin Sci (Lond) ; 124(3): 153-64, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23075334

RESUMEN

Sirtuins are members of the Sir2 (silent information regulator 2) family, a group of class III deacetylases. Mammals have seven different sirtuins, SIRT1-SIRT7. Among them, SIRT1, SIRT3 and SIRT6 are induced by calorie restriction conditions and are considered anti-aging molecules. SIRT1 has been the most extensively studied. SIRT1 deacetylates target proteins using the coenzyme NAD+ and is therefore linked to cellular energy metabolism and the redox state through multiple signalling and survival pathways. SIRT1 deficiency under various stress conditions, such as metabolic or oxidative stress or hypoxia, is implicated in the pathophysiologies of age-related diseases including diabetes, cardiovascular diseases, neurodegenerative disorders and renal diseases. In the kidneys, SIRT1 may inhibit renal cell apoptosis, inflammation and fibrosis, and may regulate lipid metabolism, autophagy, blood pressure and sodium balance. Therefore the activation of SIRT1 in the kidney may be a new therapeutic target to increase resistance to many causal factors in the development of renal diseases, including diabetic nephropathy. In addition, SIRT3 and SIRT6 are implicated in age-related disorders or longevity. In the present review, we discuss the protective functions of sirtuins and the association of sirtuins with the pathophysiology of renal diseases, including diabetic nephropathy.


Asunto(s)
Envejecimiento/metabolismo , Nefropatías Diabéticas/metabolismo , Enfermedades Renales/metabolismo , Sirtuinas/metabolismo , Animales , Restricción Calórica , Humanos , Sirtuina 1/metabolismo , Sirtuina 3/metabolismo
19.
FASEB J ; 26(7): 2963-74, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22499584

RESUMEN

This study characterizes the effect of glucose-induced activation of protein kinase Cδ (PKCδ) and Src homology-2 domain-containing phosphatase-1 (SHP-1) expression on vascular endothelial growth factor (VEGF) actions in glomerular podocytes in cultures and in glomeruli of diabetic rodents. Elevation of glucose levels induced PKCδ and p38 mitogen-activated protein kinase (p38 MAPK) to increase SHP-1 expression, increased podocyte apoptosis, and inhibited VEGF activation in podocytes and glomerular endothelial cells. The adverse effects of high glucose levels can be negated by molecular inhibitors of PKCδ, p38MAPK, and SHP-1 and only partially reduced by antioxidants and nuclear factor-κB (NF-κB) inhibitor. Increased PKCδ activation and SHP-1 expression correlated with loss of VEGF signaling and podocyte numbers in the glomeruli of diabetic rats and mice. In contrast, diabetic PKCδ-knockout (Prkcd(-/-)) mice did not exhibit activation of p38 MAPK and SHP-1 or inhibition of VEGF signaling in renal glomeruli. Functionally, diabetic Prkcd(-/-) mice had decreased expressions of TGFß, VEGF, and extracellular matrix and less albuminuria than diabetic Prkcd(+/+) mice. Hyperglycemia and diabetes can cause glomerular podocyte apoptosis and endothelial dysfunction partly due to increased PKCδ/p38 MAPK activation and the expression of SHP-1 to cause VEGF resistance, independent of NF-κB activation.


Asunto(s)
Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Secuencia de Bases , Células Cultivadas , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Células Endoteliales/metabolismo , Activación Enzimática , Femenino , Glucosa/metabolismo , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Podocitos/metabolismo , Podocitos/patología , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/deficiencia , Proteína Quinasa C-delta/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Nephrol Dial Transplant ; 28 Suppl 4: iv1-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23743020

RESUMEN

Obesity is a worldwide epidemic that is associated with several health issues, including kidney diseases. A specific kidney disease, referred to as obesity-related glomerulopathy, has been described in earlier publications. Obesity can affect the prognosis of other types of kidney diseases. Body-weight reduction with caloric restriction is an essential therapy, although strictly controlling food intake coupled with an appropriate evaluation is challenging. Low birthweight can be an important factor for obesity and results in kidney dysfunction. In this review, we analyse the consequences of obesity on kidney prognosis and potential strategies for combating obesity-associated kidney injury.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/etiología , Riñón/fisiopatología , Obesidad/complicaciones , Animales , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Humanos , Recién Nacido de Bajo Peso , Obesidad/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA