Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Circ Res ; 133(1): 71-85, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37232163

RESUMEN

BACKGROUND: As a part of natural disease progression, acute kidney injury (AKI) can develop into chronic kidney disease via renal fibrosis and inflammation. LTBP4 (latent transforming growth factor beta binding protein 4) regulates transforming growth factor beta, which plays a role in renal fibrosis pathogenesis. We previously investigated the role of LTBP4 in chronic kidney disease. Here, we examined the role of LTBP4 in AKI. METHODS: LTBP4 expression was evaluated in human renal tissues, obtained from healthy individuals and patients with AKI, using immunohistochemistry. LTBP4 was knocked down in both C57BL/6 mice and human renal proximal tubular cell line HK-2. AKI was induced in mice and HK-2 cells using ischemia-reperfusion injury and hypoxia, respectively. Mitochondrial division inhibitor 1, an inhibitor of DRP1 (dynamin-related protein 1), was used to reduce mitochondrial fragmentation. Gene and protein expression were then examined to assess inflammation and fibrosis. The results of bioenergetic studies for mitochondrial function, oxidative stress, and angiogenesis were assessed. RESULTS: LTBP4 expression was upregulated in the renal tissues of patients with AKI. Ltbp4-knockdown mice showed increased renal tissue injury and mitochondrial fragmentation after ischemia-reperfusion injury, as well as increased inflammation, oxidative stress, and fibrosis, and decreased angiogenesis. in vitro studies using HK-2 cells revealed similar results. The energy profiles of Ltbp4-deficient mice and LTBP4-deficient HK-2 cells indicated decreased ATP production. LTBP4-deficient HK-2 cells exhibited decreased mitochondrial respiration and glycolysis. Human aortic endothelial cells and human umbilical vein endothelial cells exhibited decreased angiogenesis when treated with LTBP4-knockdown conditioned media. Mitochondrial division inhibitor 1 treatment ameliorated inflammation, oxidative stress, and fibrosis in mice and decreased inflammation and oxidative stress in HK-2 cells. CONCLUSIONS: Our study is the first to demonstrate that LTBP4 deficiency increases AKI severity, consequently leading to chronic kidney disease. Potential therapies focusing on LTBP4-associated angiogenesis and LTBP4-regulated DRP1-dependent mitochondrial division are relevant to renal injury.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Daño por Reperfusión , Animales , Humanos , Ratones , Lesión Renal Aguda/prevención & control , Células Endoteliales/metabolismo , Fibrosis , Inflamación/metabolismo , Riñón/metabolismo , Proteínas de Unión a TGF-beta Latente , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Insuficiencia Renal Crónica/complicaciones , Daño por Reperfusión/complicaciones , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Factor de Crecimiento Transformador beta/metabolismo
2.
Lab Invest ; 103(7): 100146, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37004912

RESUMEN

Urokinase plasminogen activator (uPA) is a crucial activator of the fibrinolytic system that modulates tissue remodeling, cancer progression, and inflammation. However, its role in membranous nephropathy (MN) remains unclear. To clarify this issue, an established BALB/c mouse model mimicking human MN induced by cationic bovine serum albumin (cBSA), with a T helper cell type 2-prone genetic background, was used. To induce MN, cBSA was injected into Plau knockout (Plau-/-) and wild-type (WT) mice. The blood and urine samples were collected to measure biochemical parameters, such as serum concentrations of immunoglobulin (Ig)G1 and IgG2a, using enzyme-linked immunoassay. The kidneys were histologically examined for the presence of glomerular polyanions, reactive oxygen species (ROS), and apoptosis, and transmission electron microscopy was used to examine subepithelial deposits. Lymphocyte subsets were determined using flow cytometry. Four weeks post-cBSA administration, Plau-/- mice exhibited a significantly higher urine protein-to-creatine ratio, hypoalbuminemia, and hypercholesterolemia than WT mice. Histologically, compared to WT mice, Plau-/- mice showed more severe glomerular basement thickening, mesangial expansion, IgG granular deposition, intensified podocyte effacement, irregular thickening of glomerular basement membrane and subepithelial deposits, and abolishment of the glycocalyx. Moreover, increased renal ROS levels and apoptosis were observed in Plau-/- mice with MN. B-lymphocyte subsets and the IgG1-to-IgG2a ratio were significantly higher in Plau-/- mice after MN induction. Thus, uPA deficiency induces a T helper cell type 2-dominant immune response, leading to increased subepithelial deposits, ROS levels, and apoptosis in the kidneys, subsequently exacerbating MN progression in mice. This study provides a novel insight into the role of uPA in MN progression.


Asunto(s)
Glomerulonefritis Membranosa , Humanos , Animales , Ratones , Glomerulonefritis Membranosa/metabolismo , Glomerulonefritis Membranosa/patología , Albúmina Sérica Bovina/efectos adversos , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/efectos adversos , Especies Reactivas de Oxígeno , Inmunoglobulina G/efectos adversos , Inmunidad , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Colaboradores-Inductores/patología
3.
World J Clin Cases ; 10(30): 10896-10905, 2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36338217

RESUMEN

BACKGROUND: Candida esophagitis (CE) is among the commonest esophageal infections and is known as an opportunistic fungal infection mostly affecting people living with the human immunodeficiency virus (HIV). However, some medical conditions might predispose HIV-negative individuals to esophageal candidiasis. The epidemiology and associated endoscopic findings of CE among people without HIV have rarely been reported. AIM: To investigate the prevalence of CE among HIV-negative persons, and determine risk factors predicting CE. METHODS: Between January 2015 and December 2018, all consecutive outpatients who underwent routine esophagogastroduodenoscopy as part of health check-ups at their own expense at the Health Check-up Center of the Kaohsiung Veterans General Hospital, Taiwan, were recruited in this study. Those with positive HIV serology results were excluded. Sociodemographic and clinical characteristics including age, gender, economic status, smoking history, alcohol consumption, tea and coffee consumption, underlying diseases, body fat percentage, body mass index, endoscopic findings, and Helicobacter pylori infection status were carefully reviewed. CE was confirmed by endoscopic biopsy and pathological assessment with hematoxylin and eosin and periodic acid-Schiff staining. To evaluate independent factors predicting the development of CE, we conducted a univariate analysis of clinical characteristics. The variables found to be significant via univariate analysis were subsequently included in a multivariable analysis of potential risk factors for CE development. RESULTS: A total of 11802 participants were included in this study. Forty-seven (0.4%) were confirmed as having CE by pathological examination. Univariate analysis identified older age, the presence of chronic kidney disease, alcohol consumption, and steroid use (P = 0.023, < 0.001, 0.033, and 0.004, respectively) as significantly associated with CE. Multivariable analysis revealed older age [adjusted odds ratio (OR) = 1.027; 95%CI: 1.001-1.053; P = 0.045], chronic kidney disease (adjusted OR = 13.470; 95%CI: 4.574-39.673; P < 0.001), alcohol consumption (adjusted OR = 2.103; 95%CI: 1.151-3.844; P = 0.016), and steroid use (adjusted OR = 24.255; 95%CI: 5.343-110.115; P < 0.001) as independent risk factors for CE development. The presence of dysphagia was associated with severe CE (P = 0.021). CONCLUSION: The prevalence of CE among HIV-negative persons was 0.4% in Taiwan. Independent risk factors for CE were older age, chronic kidney disease, alcohol consumption, and steroid use.

4.
Int J Mol Sci ; 22(22)2021 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-34830159

RESUMEN

Medial vascular calcification has emerged as a key factor contributing to cardiovascular mortality in patients with chronic kidney disease (CKD). Vascular smooth muscle cells (VSMCs) with osteogenic transdifferentiation play a role in vascular calcification. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors reduce reactive oxygen species (ROS) production and calcified-medium-induced calcification of VSMCs. This study investigates the effects of dextromethorphan (DXM), an NADPH oxidase inhibitor, on vascular calcification. We used in vitro and in vivo studies to evaluate the effect of DXM on artery changes in the presence of hyperphosphatemia. The anti-vascular calcification effect of DXM was tested in adenine-fed Wistar rats. High-phosphate medium induced ROS production and calcification of VSMCs. DXM significantly attenuated the increase in ROS production, the decrease in ATP, and mitochondria membrane potential during the calcified-medium-induced VSMC calcification process (p < 0.05). The protective effect of DXM in calcified-medium-induced VSMC calcification was not further increased by NADPH oxidase inhibitors, indicating that NADPH oxidase mediates the effect of DXM. Furthermore, DXM decreased aortic calcification in Wistar rats with CKD. Our results suggest that treatment with DXM can attenuate vascular oxidative stress and ameliorate vascular calcification.


Asunto(s)
Dextrometorfano/farmacología , Músculo Liso Vascular , Miocitos del Músculo Liso , Estrés Oxidativo/efectos de los fármacos , Uremia , Calcificación Vascular , Animales , Línea Celular , Humanos , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Ratas , Ratas Endogámicas WKY , Uremia/tratamiento farmacológico , Uremia/metabolismo , Uremia/patología , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
5.
Cell Death Dis ; 12(10): 943, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34645813

RESUMEN

Transforming growth factor beta (TGFß) signalling regulates extracellular matrix accumulation known to be essential for the pathogenesis of renal fibrosis; latent transforming growth factor beta binding protein 4 (LTBP4) is an important regulator of TGFß activity. To date, the regulation of LTBP4 in renal fibrosis remains unknown. Herein, we report that LTBP4 is upregulated in patients with chronic kidney disease and fibrotic mice kidneys created by unilateral ureteral obstruction (UUO). Mice lacking the short LTBP4 isoform (Ltbp4S-/-) exhibited aggravated tubular interstitial fibrosis (TIF) after UUO, indicating that LTBP4 potentially protects against TIF. Transcriptomic analysis of human proximal tubule cells overexpressing LTBP4 revealed that LTBP4 influences angiogenic pathways; moreover, these cells preserved better mitochondrial respiratory functions and expressed higher vascular endothelial growth factor A (VEGFA) compared to wild-type cells under hypoxia. Results of the tube formation assay revealed that additional LTBP4 in human umbilical vein endothelial cell supernatant stimulates angiogenesis with upregulated vascular endothelial growth factor receptors (VEGFRs). In vivo, aberrant angiogenesis, abnormal mitochondrial morphology and enhanced oxidative stress were observed in Ltbp4S-/- mice after UUO. These results reveal novel molecular functions of LTBP4 stimulating angiogenesis and potentially impacting mitochondrial structure and function. Collectively, our findings indicate that LTBP4 protects against disease progression and may be of therapeutic use in renal fibrosis.


Asunto(s)
Riñón/patología , Proteínas de Unión a TGF-beta Latente/metabolismo , Mitocondrias/ultraestructura , Neovascularización Fisiológica , Animales , Diferenciación Celular , Medios de Cultivo Condicionados/farmacología , Fibrosis , Perfilación de la Expresión Génica , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunidad , Túbulos Renales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Modelos Biológicos , Fenotipo , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología
6.
Cancer Lett ; 499: 290-300, 2021 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-33271263

RESUMEN

Protocadherin 10 (PCDH10) is identified as a tumor suppressor in multiple cancers. The molecular mechanisms that mediate the functions of PCDH10 have yet to be fully elucidated. Here, we demonstrated that ectopic expression of PCDH10 in colorectal cancer (CRC) cells induced cell cycle retardation and increased apoptosis through regulation of the p53/p21/Rb axis and Bcl-2 expression. Overexpression of PCDH10 reversed the epithelial-mesenchymal transition (EMT) process with morphological changes and EMT marker alterations. Mechanistic study revealed that PCDH10 inhibited AKT/GSK3ß signaling pathway which in turn reduced ß-catenin activity and thus attenuated Snail and Twist1 expression. Furthermore, PCDH10 inhibited the stemness of CRC cells, including spheroid formation and stem cell markers. A proteomics approach revealed that PCDH10 could interact with EGFR, which was further verified by co-immunoprecipitation. Moreover, restoration of PCDH10 expression reduced EGFR phosphorylation. Accordingly, our work proposes a novel pathway by which PCDH10 directly engages in the negative regulation of EGFR/AKT/ß-catenin signaling pathway, resulting in tumor suppression.


Asunto(s)
Cadherinas/metabolismo , Neoplasias Colorrectales/patología , Proteínas Supresoras de Tumor/metabolismo , Apoptosis , Cadherinas/genética , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Protocadherinas , Esferoides Celulares , Proteínas Supresoras de Tumor/genética , Vía de Señalización Wnt
7.
Cell Rep ; 29(5): 1261-1273.e6, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31665638

RESUMEN

Acute kidney injury (AKI) is characterized by mitochondrial dysfunction and activation of the innate immune system. The cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) pathway detects cytosolic DNA and induces innate immunity. Here, we investigate the role of mitochondrial damage and subsequent activation of the cGAS-STING pathway using a genetically engineered animal model of cisplatin-induced AKI and cultured tubular cells. Cisplatin induced mtDNA leakage into the cytosol-probably through BCL-2-like protein 4 (BAX) pores in the mitochondrial outer membrane-in tubules, with subsequent activation of the cGAS-STING pathway, thereby triggering inflammation and AKI progression, which is improved in STING-deficient mice. STING knockdown in cultured tubular cells ameliorates inflammatory responses induced by cisplatin. mtDNA depletion and repletion studies support tubular inflammatory responses via the cGAS-STING signal activation by cytosolic mtDNA. Therefore, we conclude that mitochondrial dysfunction and subsequent activation of the mtDNA-cGAS-STING pathway is a critical regulator of kidney injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Inflamación/patología , Proteínas de la Membrana/metabolismo , Mitocondrias/patología , Nucleotidiltransferasas/metabolismo , Lesión Renal Aguda/inducido químicamente , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Cisplatino/efectos adversos , Citosol/metabolismo , ADN Mitocondrial/metabolismo , Humanos , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
8.
Kidney Int ; 95(3): 577-589, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30639234

RESUMEN

Tubulointerstitial fibrosis is a strong predictor of progression in patients with chronic kidney disease, and is often accompanied by lipid accumulation in renal tubules. However, the molecular mechanisms modulating the relationship between lipotoxicity and tubulointerstitial fibrosis remain obscure. ATF6α, a transcription factor of the unfolded protein response, is reported to be an upstream regulator of fatty acid metabolism. Owing to their high energy demand, proximal tubular cells (PTCs) use fatty acids as their main energy source. We therefore hypothesized that ATF6α regulates PTC fatty acid metabolism, contributing to lipotoxicity-induced tubulointerstitial fibrosis. Overexpression of activated ATF6α transcriptionally downregulated peroxisome proliferator-activated receptor-α (PPARα), the master regulator of lipid metabolism, leading to reduced activity of fatty acid ß-oxidation and cytosolic accumulation of lipid droplets in a human PTC line (HK-2). ATF6α-induced lipid accumulation caused mitochondrial dysfunction, enhanced apoptosis, and increased expression of connective tissue growth factor (CTGF), as well as reduced cell viability. Atf6α-/- mice had sustained expression of PPARα and less tubular lipid accumulation following unilateral ischemia-reperfusion injury (uIRI), resulting in the amelioration of apoptosis; reduced expression of CTGF, α-smooth muscle actin, and collagen I; and less tubulointerstitial fibrosis. Administration of fenofibrate, a PPARα agonist, reduced lipid accumulation and tubulointerstitial fibrosis in the uIRI model. Taken together, these findings suggest that ATF6α deranges fatty acid metabolism in PTCs, which leads to lipotoxicity-mediated apoptosis and CTGF upregulation, both of which promote tubulointerstitial fibrosis.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Túbulos Renales Proximales/patología , PPAR alfa/metabolismo , Insuficiencia Renal Crónica/patología , Factor de Transcripción Activador 6/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ácidos Grasos/metabolismo , Fibrosis , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Insuficiencia Renal Crónica/etiología
9.
Kidney Int ; 94(3): 536-550, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29887316

RESUMEN

Lipotoxicity plays an important role in the progression of chronic kidney damage via various mechanisms, such as endoplasmic reticulum stress. Several studies proposed renal lipotoxicity in glomerular and tubular cells but the effect of lipid on renal erythropoietin (EPO)-producing (REP) cells in the interstitium has not been elucidated. Since renal anemia is caused by derangement of EPO production in REP cells, we evaluated the effect of palmitate, a representative long-chain saturated fatty acid, on EPO production and the endoplasmic reticulum stress pathway. EPO production was suppressed by palmitate (palmitate-conjugated bovine serum albumin [BSA]) or a high palmitate diet, but not oleic acid-conjugated BSA or a high oleic acid diet, especially under cobalt-induced pseudo-hypoxia both in vitro and in vivo. Importantly, suppression of EPO production was not induced by a decrease in transcription factor HIF activity, while it was significantly associated with endoplasmic reticulum stress, particularly transcription factor ATF4 activation, which suppresses 3'-enhancer activity of the EPO gene. ATF4 knockdown by siRNA significantly attenuated the suppressive effect of palmitate on EPO production. Studies utilizing inherited super-anemic mice (ISAM) mated with EPO-Cre mice (ISAM-REC mice) for lineage-labeling of REP cells showed that ATF4 activation by palmitate suppressed EPO production in REP cells. Laser capture microdissection confirmed ATF4 activation in the interstitial area of ISAM-REC mice treated with palmitate-conjugated BSA. Thus, endoplasmic reticulum stress induced by palmitate suppressed EPO expression by REP cells in a manner independent of HIF activation. The link between endoplasmic reticulum stress, dyslipidemia, and hypoxia may contribute to development and progression of anemia in CKD.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Anemia/patología , Eritropoyetina/metabolismo , Riñón/metabolismo , Palmitatos/metabolismo , Factor de Transcripción Activador 4/genética , Anemia/sangre , Anemia/etiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Eritropoyetina/sangre , Eritropoyetina/genética , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Captura por Microdisección con Láser , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/metabolismo , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología , Respuesta de Proteína Desplegada
10.
Sci Rep ; 7(1): 11168, 2017 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-28894140

RESUMEN

The prevalence of chronic kidney disease (CKD), characterized by progressive renal dysfunction with tubulointerstitial fibrosis, is increasing because of societal aging. Uremic toxins, accumulated during renal dysfunction, cause kidney damage, leading to renal deterioration. A recent metabolomic analysis revealed that plasma D-serine accumulation is associated with faster progression of renal dysfunction in CKD patients. However, the causal relationship and the underlying mechanisms remain unclear. Herein, we demonstrated that D-serine markedly induced cellular senescence and apoptosis in a human proximal tubular cell line, HK-2, and primary culture of human renal tubular cells. The former was accompanied by G2/M cell cycle arrest and senescence-associated secretory phenotype, including pro-fibrotic and pro-inflammatory factors, contributing to tubulointerstitial fibrosis. Integrated stress response mediated by the general control nonderepressible 2 played an important role in D-serine-induced tubular cell toxicity and pro-fibrotic phenotypes, accelerating CKD progression and kidney aging. D-serine upregulated the L-serine synthesis pathway. Furthermore, D-serine-induced suppression of tubular cell proliferation was ameliorated by L-serine administration, indicating that D-serine exposure induced an L-serine-deprived state in tubular cells, compensated by L-serine synthesis. Thus, this study unveils molecular mechanisms underlying D-serine-induced tubular damage and pro-fibrotic phenotypes, suggesting that D-serine is a uremic toxin involved in CKD pathogenesis.


Asunto(s)
Envejecimiento/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Enfermedades Renales/fisiopatología , Túbulos Renales Proximales/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Serina/metabolismo , Apoptosis , Puntos de Control del Ciclo Celular , Células Cultivadas , Células Epiteliales/patología , Humanos , Túbulos Renales Proximales/patología
11.
Kidney Int ; 92(4): 782-784, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28938947

RESUMEN

Podocyte biology is an important key factor for the maintenance of glomerular function and structure. Nephronectin is a podocyte-derived extracellular matrix protein in the glomerular basement membrane. Suppression of nephronectin expression by microRNA-378a-3p significantly induced proteinuria due to loss of integrity of the glomerular filtration barrier in zebrafish and mice, demonstrating the relevance of the microRNA-nephronectin axis in regulation of the podocyte-glomerular basement membrane interaction and integrity of the glomerular filtration barrier.


Asunto(s)
MicroARNs , Podocitos , Animales , Proteínas de la Matriz Extracelular , Membrana Basal Glomerular , Ratones , Proteinuria
12.
Clin Exp Pharmacol Physiol ; 44 Suppl 1: 70-77, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28467603

RESUMEN

Chronic kidney disease (CKD) is a major concern in public health. The pathology of CKD includes premature ageing in the kidney and vessels, which results in a high risk of cardiovascular events and end-stage renal disease. Many factors are involved in premature ageing in CKD, including hormonal imbalance, glycative stress, nitrogenous metabolites, and oxidative stress. Of these, the most important role in premature ageing in CKD is played by glycative stress, namely a massive and unfavourable glycation state, since the kidney is responsible for the clearance of advanced glycation endproducts (AGEs). In an animal model, overexpression of glyoxalase I (GLO-1), a detoxifier of AGEs, has been found to alleviate premature ageing in the kidney and vessels. Both lifestyle changes and drug therapy have shown promise in overcoming premature ageing. Promising drug therapies include a GLO-1 activator and an absorbent against glycotoxin and nitrogenous metabolites.


Asunto(s)
Envejecimiento Prematuro , Senescencia Celular , Productos Finales de Glicación Avanzada/metabolismo , Riñón/metabolismo , Insuficiencia Renal Crónica/metabolismo , Hipoxia de la Célula , Senescencia Celular/efectos de los fármacos , Activación Enzimática , Activadores de Enzimas/uso terapéutico , Fragilidad/metabolismo , Fragilidad/fisiopatología , Estado de Salud , Hormonas/metabolismo , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Lactoilglutatión Liasa/metabolismo , Estrés Oxidativo , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/terapia , Conducta de Reducción del Riesgo , Transducción de Señal , Equilibrio Hidroelectrolítico
13.
Nutrients ; 9(4)2017 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-28375181

RESUMEN

Dietary contents and their metabolites are closely related to chronic kidney disease (CKD) progression. Advanced glycated end products (AGEs) are a type of uremic toxin produced by glycation. AGE accumulation is not only the result of elevated glucose levels or reduced renal clearance capacity, but it also promotes CKD progression. Indoxyl sulfate, another uremic toxin derived from amino acid metabolism, accumulates as CKD progresses and induces tubulointerstitial fibrosis and glomerular sclerosis. Specific types of amino acids (d-serine) or fatty acids (palmitate) are reported to be closely associated with CKD progression. Promising therapeutic targets associated with nutrition include uremic toxin absorbents and inhibitors of AGEs or the receptor for AGEs (RAGE). Probiotics and prebiotics maintain gut flora balance and also prevent CKD progression by enhancing gut barriers and reducing uremic toxin formation. Nrf2 signaling not only ameliorates oxidative stress but also reduces elevated AGE levels. Bardoxolone methyl, an Nrf2 activator and NF-κB suppressor, has been tested as a therapeutic agent, but the phase 3 clinical trial was terminated owing to the high rate of cardiovascular events. However, a phase 2 trial has been initiated in Japan, and the preliminary analysis reveals promising results without an increase in cardiovascular events.


Asunto(s)
Dieta , Insuficiencia Renal Crónica/epidemiología , Acidosis/epidemiología , Aminoácidos/efectos adversos , Metabolismo de los Hidratos de Carbono , Carbohidratos de la Dieta/efectos adversos , Ácidos Grasos/efectos adversos , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Prebióticos/administración & dosificación , Probióticos/administración & dosificación , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Insuficiencia Renal Crónica/prevención & control
14.
Oncotarget ; 7(51): 84938-84950, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27793051

RESUMEN

Glucosaminyl N-deacetylase/N-sulfotransferases (NDSTs) are the first enzymes that mediate the initiation of heparan sulfate sulfation. We previously identified NDST4 as a putative tumor suppressor in human colorectal cancer. In the study, we generated an Ndst4 knockout (Ndst4-/-) mouse strain and explored its phenotypic characteristics, particularly in the development of colonic epithelial homeostasis. The Ndst4-deficient mice were viable and fertile, and their life spans were similar to those of wild-type littermates. No gross behavioral or morphological differences were observed between the Ndst4-/- and wild-type mice, and no significant changes were determined in the hematological or serum biochemical parameters of the Ndst4-/- mice. Ndst4 RNA transcripts were expressed in the brain, lung, gastrointestinal tract, pancreas, and ovary. However, Ndst4-null mice exhibited no gross or histological abnormalities in the studied organs, except for the colon. Although no alterations were observed in the crypt length or number of proliferating cells, the Ndst4-/- mice exhibited an increased number of goblet cells and a decreased number of colonocytes in the proximal colon compared with the wild-type mice. Moreover, Ndst4 deficiency increased the basal level of apoptosis in the colonic epithelium. Taken together, we established, for the first time, an Ndst4-/- mouse strain and revealed the involvement of Ndst4 in the development and homeostasis of colonic epithelium. Accordingly, NDST4 in human colon might direct the biosynthesis of specific heparan sulfate proteoglycans that are essential for the maintenance of colonic epithelial homeostasis. Thus, the loss of its function may result in the tumorigenesis and progression of colorectal cancer.


Asunto(s)
Colon/patología , Neoplasias Colorrectales/metabolismo , Células Epiteliales/fisiología , Células Caliciformes/fisiología , Sulfotransferasas/metabolismo , Animales , Apoptosis , Carcinogénesis , Células Cultivadas , Colon/fisiología , Neoplasias Colorrectales/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sulfotransferasas/genética
15.
Ann Surg Oncol ; 22 Suppl 3: S1481-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25968618

RESUMEN

BACKGROUND: Shisa3 is a novel tumor suppressor identified in lung cancer. However, its antitumor activity in other human cancers and the mechanism of gene inactivation remain unknown. METHODS: SHISA3 expression was measured by reverse transcription-PCR (RT-PCR) and quantitative RT-PCR (RT-qPCR). DNA methylation was determined by bisulfite sequencing and pyrosequencing. RESULTS: Down-regulation of SHISA3 expression was observed in all of 11 colorectal cancer (CRC) cell lines and was further confirmed in 34 (65.4 %) of 52 colorectal carcinomas by RT-qPCR. Four of six CRC cell lines could restore SHISA3 expression after treatment with 5-aza-2'-deoxycytidine. Tumor-specific methylation of five CpG sites in the first intron of SHISA3 was identified by bisulfite sequencing, and their methylation levels were quantified in 127 pairs of primary CRC tissues by bisulfite pyrosequencing. The methylation levels of SHISA3 in tumors were noticeably higher than that in their matched normal mucosae. In addition, SHISA3 hypermethylation was significantly associated with an increased risk of disease recurrence in patients with stage II and III disease (P = 0.007) and was an independent predictor of poor overall survival [hazard ratio (HR) 2.9, 95 % confidence interval (CI) 1.5-5.8; P = 0.002] and disease-free survival (HR 4.0, 95 % CI 1.6-10.2; P = 0.003) of CRC patients. CONCLUSIONS: SHISA3 gene is epigenetically inactivated in a substantial fraction of CRC, and its hypermethylation is of prognostic significance in predicting clinical outcome. The quantitative bisulfite pyrosequencing assay established could be a cost-effective tool for providing a potential biomarker of adverse prognosis in CRC.


Asunto(s)
Adenocarcinoma/genética , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Metilación de ADN , Proteínas de la Membrana/genética , Recurrencia Local de Neoplasia/genética , Adenocarcinoma/secundario , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Neoplasias Colorrectales/patología , Islas de CpG , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfitos , Tasa de Supervivencia , Células Tumorales Cultivadas
16.
Int J Cancer ; 135(11): 2593-603, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24740680

RESUMEN

Protocadherin 10 (PCDH10), a novel tumor suppressor gene in human cancers, is located in a common deleted region at chromosome 4q28 in colorectal cancer (CRC). This study aimed to ascertain the genetic loss of PCDH10 and its clinical relevance in CRC and to explore the tumor suppressor function of PCDH10. The genetic deletion of PCDH10 was determined in 171 pairs of primary tumors and corresponding normal mucosae by loss of heterozygosity study. In total, 53 carcinomas were positive for allelic loss of PCDH10. The genetic aberration was significantly associated with tumor progression and distant metastasis (p = 0.021 and p = 0.018, respectively) and was an independent predictor of poor survival for CRC patients (p = 0.005). Expression of PCDH10 gene was silenced or markedly down-regulated in all of 12 CRC cell lines tested and in 41 of 53 colorectal carcinomas compared with their matched normal mucosae. Ectopic expression of PCDH10 suppressed cancer cell proliferation, anchorage-independent growth, migration and invasion in vitro. Subcutaneous injection of PCDH10-expressing CRC cells into SCID mice revealed the reduction of tumor growth compared with that observed in mock-inoculated mice. Furthermore, through intrasplenic implantation, the re-expression of PCDH10 in silenced cells restrained liver metastasis and improved survival in SCID mice. In conclusion, PCDH10 is a pivotal tumor suppressor in CRC, and the loss of its function promotes not only tumor progression but also liver metastasis. In addition, the genetic deletion of PCDH10 represents an adverse prognostic marker for the survival of patients with CRC.


Asunto(s)
Cadherinas/genética , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/prevención & control , Neoplasias Hepáticas/prevención & control , Pérdida de Heterocigocidad , Recurrencia Local de Neoplasia/prevención & control , Animales , Apoptosis , Western Blotting , Cadherinas/metabolismo , Adhesión Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Femenino , Citometría de Flujo , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Técnicas para Inmunoenzimas , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/secundario , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Protocadherinas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas
17.
PLoS One ; 8(6): e67040, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825612

RESUMEN

BACKGROUND: Genomic deletion at tumor suppressor loci is a common genetic aberration in human cancers. The study aimed to explore candidate tumor suppressor genes at chromosome 4q25-q28.2 and to delineate novel prognostic biomarkers associated with colorectal cancer (CRC). METHODS: Deletion mapping of chromosome 4q25-q28.2 was conducted in 114 sporadic CRC by loss of heterozygosity study with 11 microsatellite markers. A novel candidate tumor suppressor gene, namely NDST4, was identified at 4q26. Gene expression of NDST4 was investigated in 52 pairs of primary CRC tissues by quantitative reverse transcription-polymerase chain reaction. Allelic loss of NDST4 gene was further determined in 174 colorectal carcinomas by loss of heterozygosity analysis, and then was assessed for clinical relevance. RESULTS: One minimal deletion region was delineated between D4S2297 and D4S2303 loci at 4q26, where NDST4 was the only gene that had markedly been downregulated in CRC tumors. By laser capture microdissection, NDST4 RNA expression was demonstrated in colonic epithelial cells, but was undetectable in tumor cells. In total, 30 (57.7%) of 52 colorectal carcinomas showed a dramatic reduction in NDST4 gene expression compared with matched normal mucosae. The genetic loss of NDST4 was significantly associated with advanced pathological stage (P = 0.039) and poorer overall survival of patients (P = 0.036). CONCLUSIONS: NDST4 gene is a novel candidate tumor suppressor gene in human cancer, and the loss of its function might be involved in CRC progression. In addition, the loss of heterozygosity assay, which was established to determine the allelic loss of NDST4 gene, could be a cost-effective tool for providing a useful biomarker of adverse prognosis in CRC.


Asunto(s)
Cromosomas Humanos Par 4/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Genes Supresores de Tumor , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Sulfotransferasas/deficiencia , Sulfotransferasas/genética , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Pólipos del Colon/genética , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Sitios Genéticos/genética , Humanos , Mucosa Intestinal/metabolismo , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico
18.
J Virol ; 85(6): 2927-41, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21228244

RESUMEN

Reverse genetics is a powerful tool to study single-stranded RNA viruses. Despite tremendous efforts having been made to improve the methodology for constructing flavivirus cDNAs, the cause of toxicity of flavivirus cDNAs in bacteria remains unknown. Here we performed mutational analysis studies to identify Escherichia coli promoter (ECP) sequences within nucleotides (nt) 1 to 3000 of the dengue virus type 2 (DENV2) and Japanese encephalitis virus (JEV) genomes. Eight and four active ECPs were demonstrated within nt 1 to 3000 of the DENV2 and JEV genomes, respectively, using fusion constructs containing DENV2 or JEV segments and empty vector reporter gene Renilla luciferase. Full-length DENV2 and JEV cDNAs were obtained by inserting mutations reducing their ECP activity in bacteria without altering amino acid sequences. A severe cytopathic effect occurred when BHK21 cells were transfected with in vitro-transcribed RNAs from either a DENV2 cDNA clone with multiple silent mutations within the prM-E-NS1 region of dengue genome or a JEV cDNA clone with an A-to-C mutation at nt 90 of the JEV genome. The virions derived from the DENV2 or JEV cDNA clone exhibited infectivities similar to those of their parental viruses in C6/36 and BHK21 cells. A cis-acting element essential for virus replication was revealed by introducing silent mutations into the central portion (nt 160 to 243) of the core gene of DENV2 infectious cDNA or a subgenomic DENV2 replicon clone. This novel strategy of constructing DENV2 and JEV infectious clones could be applied to other flaviviruses or pathogenic RNA viruses to facilitate research in virology, viral pathogenesis, and vaccine development.


Asunto(s)
ADN Complementario , ADN Viral , Virus del Dengue/crecimiento & desarrollo , Virus de la Encefalitis Japonesa (Especie)/crecimiento & desarrollo , Regiones Promotoras Genéticas , Virología/métodos , Replicación Viral , Animales , Fusión Artificial Génica , Línea Celular , Cricetinae , Efecto Citopatogénico Viral , Virus del Dengue/genética , Virus de la Encefalitis Japonesa (Especie)/genética , Escherichia coli/genética , Genes Reporteros , Ingeniería Genética/métodos , Luciferasas de Renilla/genética , Luciferasas de Renilla/metabolismo , Mutación , Recombinación Genética , Saccharomyces cerevisiae/genética , Cultivo de Virus/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA