Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Curr Opin Nephrol Hypertens ; 31(1): 129-134, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34710887

RESUMEN

PURPOSE OF REVIEW: Diabetic kidney disease (DKD) is a significant health concern. Innovative strategies to prevent or limit the progression of DKD are urgently needed due to the limitation of existing treatments. KCa3.1, a potassium channel, is involved in a range of biological processes from cell survival to cell death. This review summarizes the current knowledge on the pathophysiological functions of the KCa3.1 channel, specifically its involvement in maintaining mitochondrial function. More specifically, the therapeutic potential of targeting KCa3.1 in DKD is systematically discussed in the review. RECENT FINDINGS: Mitochondrial dysfunction contributes to the development and progression of DKD. Accumulating evidence indicates that KCa3.1 dysregulation plays a crucial role in mitochondrial dysfunction, in addition to driving cellular activation, proliferation and inflammation. Recent studies demonstrate that KCa3.1 deficiency improves diabetes-induced mitochondrial dysfunction in DKD, which is attributed to modulation of mitochondrial quality control through mitigating the altered mitochondrial dynamics and restoring abnormal BNIP3-mediated mitophagy. SUMMARY: Based on its role in fibrosis, inflammation and mitochondrial dysfunction, pharmacological inhibition of KCa3.1 may offer a promising alternative for the treatment of DKD. Due to its safety profile in humans, the repurposing of senicapoc has the potential to expedite an urgently needed new drug in DKD.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/etiología , Fibrosis , Humanos , Inflamación , Mitocondrias
2.
Int J Mol Sci ; 23(14)2022 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-35887386

RESUMEN

Chronic kidney disease (CKD) is a growing global public health problem. The implementation of evidence-based clinical practices only defers the development of kidney failure. Death, transplantation, or dialysis are the consequences of kidney failure, resulting in a significant burden on the health system. Hence, innovative therapeutic strategies are urgently needed due to the limitations of current interventions. Photobiomodulation (PBM), a form of non-thermal light therapy, effectively mitigates mitochondrial dysfunction, reactive oxidative stress, inflammation, and gut microbiota dysbiosis, all of which are inherent in CKD. Preliminary studies suggest the benefits of PBM in multiple diseases, including CKD. Hence, this review will provide a concise summary of the underlying action mechanisms of PBM and its potential therapeutic effects on CKD. Based on the findings, PBM may represent a novel, non-invasive and non-pharmacological therapy for CKD, although more studies are necessary before PBM can be widely recommended.


Asunto(s)
Microbioma Gastrointestinal , Terapia por Luz de Baja Intensidad , Insuficiencia Renal Crónica , Disbiosis , Humanos , Inflamación , Diálisis Renal , Insuficiencia Renal Crónica/radioterapia
3.
Int J Mol Sci ; 22(12)2021 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-34202940

RESUMEN

Acute kidney injury (AKI) and chronic kidney disease (CKD) are rising in global prevalence and cause significant morbidity for patients. Current treatments are limited to slowing instead of stabilising or reversing disease progression. In this review, we describe mesenchymal stem cells (MSCs) and their constituents, extracellular vesicles (EVs) as being a novel therapeutic for CKD. MSC-derived EVs (MSC-EVs) are membrane-enclosed particles, including exosomes, which carry genetic information that mimics the phenotype of their cell of origin. MSC-EVs deliver their cargo of mRNA, miRNA, cytokines, and growth factors to target cells as a form of paracrine communication. This genetically reprograms pathophysiological pathways, which are upregulated in renal failure. Since the method of exosome preparation significantly affects the quality and function of MSC-exosomes, this review compares the methodologies for isolating exosomes from MSCs and their role in tissue regeneration. More specifically, it summarises the therapeutic efficacy of MSC-EVs in 60 preclinical animal models of AKI and CKD and the cargo of biomolecules they deliver. MSC-EVs promote tubular proliferation and angiogenesis, and inhibit apoptosis, oxidative stress, inflammation, the epithelial-to-mesenchymal transition, and fibrosis, to alleviate AKI and CKD. By reprogramming these pathophysiological pathways, MSC-EVs can slow or even reverse the progression of AKI to CKD, and therefore offer potential to transform clinical practice.


Asunto(s)
Terapia Biológica , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Enfermedades Renales/terapia , Células Madre Mesenquimatosas/metabolismo , Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/terapia , Animales , Apoptosis/efectos de los fármacos , Terapia Biológica/métodos , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células , Fraccionamiento Químico , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Exosomas/metabolismo , Humanos , Enfermedades Renales/etiología , Enfermedades Renales/patología , Células Madre Mesenquimatosas/citología , Sustancias Protectoras , Insuficiencia Renal/diagnóstico , Insuficiencia Renal/etiología , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/terapia
4.
Int J Mol Sci ; 22(4)2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-33546409

RESUMEN

Mitochondria are critical organelles that play a key role in cellular metabolism, survival, and homeostasis. Mitochondrial dysfunction has been implicated in the pathogenesis of diabetic kidney disease. The function of mitochondria is critically regulated by several mitochondrial protein kinases, including the phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1). The focus of PINK1 research has been centered on neuronal diseases. Recent studies have revealed a close link between PINK1 and many other diseases including kidney diseases. This review will provide a concise summary of PINK1 and its regulation of mitochondrial function in health and disease. The physiological role of PINK1 in the major cells involved in diabetic kidney disease including proximal tubular cells and podocytes will also be summarized. Collectively, these studies suggested that targeting PINK1 may offer a promising alternative for the treatment of diabetic kidney disease.


Asunto(s)
Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Susceptibilidad a Enfermedades , Mitocondrias/enzimología , Proteínas Quinasas/metabolismo , Animales , Autofagia , Activación Enzimática , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Túbulos Renales/metabolismo , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Podocitos/metabolismo , Proteínas Quinasas/genética
5.
J Cell Physiol ; 233(9): 7045-7054, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29380373

RESUMEN

Progressive tubulointerstitial fibrosis has been recognized as a common pathological process that leads to the progression of all chronic kidney disease (CKD). Innovative strategies are needed to both prevent and treat CKD. Inflammatory and fibrotic signaling pathways play central roles in the progression of CKD regardless of aetiology. Hence, targeting inflammatory and fibrotic responses holds promise to limit renal fibrosis. Metformin has been the most prescribed glucose-lowering medicine worldwide, and its potential for many other therapeutic applications is also being explored intensively. Increasing evidence indicates metformin may limit renal fibrosis. However, the exact mechanisms whereby metformin limits renal injury are not fully understood. The anti-fibrotic effects of metformin, independent of improved glycaemic control was examined in a folic acid-induced mouse model of nephropathy for 14 days. Human proximal tubular cells (HK2 cells) exposed to TGF-ß1 were used in in vitro models to examine mechanistic pathways. Folic acid induced nephropathy was associated with the overexpression of inflammatory markers MCP-1, F4/80, type IV collagen, fibronectin and TGF-ß1 compared to control groups, which were partially attenuated by metformin treatment. In vitro studies confirmed that metformin inhibited TGF-ß1 induced inflammatory and fibrotic responses through Smad3, ERK1/2, and P38 pathways in human renal proximal tubular cells. These results suggest that metoformin attenuates folic acid-induced renal interstitial fibrogenesis through TGF-ß1 signaling pathways.


Asunto(s)
Riñón/patología , Metformina/farmacología , Albuminuria/complicaciones , Albuminuria/patología , Animales , Línea Celular , Quimiocina CCL2/metabolismo , Colágeno Tipo IV/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Fibronectinas/metabolismo , Fibrosis , Ácido Fólico , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/patología , Riñón/efectos de los fármacos , Riñón/fisiopatología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
6.
Biomacromolecules ; 17(3): 965-73, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26807678

RESUMEN

We present the preparation of 11 nm polyacrylamide-stabilized polystyrene latex particles for conjugation to a microRNA model by surfactant-free RAFT emulsion polymerization. Our synthetic strategy involved the preparation of amphiphilic polyacrylamide-block-polystyrene copolymers, which were able to self-assemble into polymeric micelles and "grow" into polystyrene latex particles. The surface of these sterically stabilized particles was postmodified with a disulfide-bearing linker for the attachment of the microRNA model, which can be released from the latex particles under reducing conditions. These nanoparticles offer the advantage of ease of preparation via a scaleable process, and the versatility of their synthesis makes them adaptable to a range of applications.


Asunto(s)
Portadores de Fármacos/síntesis química , Látex/química , MicroARNs/administración & dosificación , Nanopartículas/química , Poliestirenos/química , Resinas Acrílicas/química , Portadores de Fármacos/química , Liberación de Fármacos , Emulsiones/química , Oxidación-Reducción , Polimerizacion , Tensoactivos/química
7.
Curr Opin Nephrol Hypertens ; 24(1): 61-6, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25415613

RESUMEN

PURPOSE OF REVIEW: Hypertension and hyperglycaemia are major risk factors that result in chronic kidney disease (CKD). Achievement of blood pressure goals, optimal control of blood glucose levels and the use of agents to block the renin-angiotensin-aldosterone system slow the progression of CKD. However, not all patients are benefited by these interventions and novel strategies to arrest or reverse the pathological processes inherent in CKD are needed. The therapeutic potential of targeting KCa3.1 in CKD will be discussed in this review. RECENT FINDINGS: Blockade of KCa3.1 ameliorates activation of renal fibroblasts in diabetic mice by inhibiting the transforming growth factor-ß1/small mothers against decapentaplegic pathway. A concomitant reduction in nuclear factor-κB activation in human proximal tubular cells under diabetic conditions has been observed. Advanced glycosylated endproducts induce both protein expression and current density of KCa3.1, which, in turn, mediates migration and proliferation of vascular smooth muscle cells via Ca²âº-dependent signalling pathways. SUMMARY: Studies have clearly demonstrated a causal role of chronic hyperglycaemia and hypertension in the development of CKD. However, a large proportion of patients develop end-stage kidney disease despite strict glycaemic control and the attainment of recommended blood pressure goals. Therefore, it is essential to identify and validate novel targets to reduce the development and progression of CKD. Recent findings demonstrate that genetic deletion or pharmacologic inhibition of KCa3.1 significantly reduces the development of diabetic nephropathy in animal models. However, the consequences of blockade of KCa3.1 in preventing and treating established diabetic nephropathy in humans warrants further study.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Insuficiencia Renal Crónica/tratamiento farmacológico , Movimiento Celular , Proliferación Celular , Nefropatías Diabéticas/metabolismo , Fibroblastos/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Hipertensión/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Músculo Liso Vascular/fisiología , FN-kappa B/metabolismo , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/prevención & control , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
8.
Clin Exp Pharmacol Physiol ; 42(6): 680-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25882815

RESUMEN

Krüppel-like factor 4 (KLF4) is known to mitigate inflammation in several cell types. Using human proximal tubule cells, the present study aimed to investigate the role of KLF4 in regulating transforming growth factor (TGF)-ß1 induced inflammatory and fibrotic responses. Human kidney proximal tubular cells were exposed to high glucose, or TGF-ß1 and KLF4 expressions were determined. Cells were then transfected with empty vector or KLF4 and exposed to 2-ng/mL TGF-ß1 for up to 72 h. Inflammatory proteins (macrophage migration inhibitory factor and monocyte chemoattractant protein-1) and pro-fibrotic proteins (fibronectin and collagen IV) were measured after 72 h by enzyme-linked immunosorbent assay and western blot, respectively. To determine the relevance to in vivo models of chronic kidney disease, KLF4 protein expression in streptozotocin-induced diabetic mice was determined. Krüppel-like factor 4 messenger RNA (mRNA) levels were significantly reduced in high glucose-treated human kidney proximal tubular cells. High glucose increased TGF-ß1 mRNA expression, which significantly increased migration inhibitory factor and monocyte chemoattractant protein-1 protein secretion. Transforming growth factor-ß1 significantly increased fibronectin and collagen IV protein expression. The overexpression of KLF4 significantly reduced TGF-ß-mediated increases in migration inhibitory factor and monocyte chemoattractant protein-1 but had no effect on TGF-ß-mediated fibronectin and collagen IV mRNA and protein expression. The levels of KLF4 mRNA were significantly reduced in the diabetic kidney, and diabetic animals had a significant reduction in renal tubular expression of KLF4 proteins. This data suggest that KLF4 reduces inflammation induced by TGF-ß1, suggesting a therapeutic role for KLF4 in diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Túbulos Renales Proximales/metabolismo , Factores de Transcripción de Tipo Kruppel/fisiología , Factor de Crecimiento Transformador beta1/toxicidad , Animales , Células Cultivadas , Diabetes Mellitus Experimental/patología , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Mediadores de Inflamación/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Factor 4 Similar a Kruppel , Ratones , Ratones Noqueados
9.
Lab Invest ; 94(3): 309-20, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24492284

RESUMEN

Thioredoxin-interacting protein (TXNIP) expression is ubiquitous and is induced by a variety of cellular stresses, including high intracellular glucose. TXNIP is associated with activation of oxidative stress and tubulointerstitial fibrosis in diabetic nephropathy. Autophagy is a major pathway that delivers damaged proteins and organelles to lysosomes to maintain cellular homeostasis. This study aimed to investigate the dysregulation of autophagy and the regulation of TXNIP on autophagy in renal proximal tubular cells (PTCs) under diabetic conditions. The formation of autophagosomes was measured using transmission electron microscopy, and LC3-II, and the effectiveness of autophagic clearance was determined by p62 expression in diabetic kidney and in human PTCs exposed to high glucose (HG). The results collectively demonstrated increased expression of TXNIP, LC3/LC3-II and p62 in renal tubular cells of mice with diabetic nephropathy and in cultured human PTCs exposed to HG (30 mM/l) for 48 h compared with control. The formation of autophagic vacuoles was increased in HG-induced cells. Furthermore, silencing of TXNIP by siRNA transfection reduced autophagic vacuoles and the expression of LC3-II and p62 in human PTCs exposed to HG compared with control and partially reversed the accumulation of LC3-II and p62 induced by bafilomycin A1 (50 nM/l), a pharmacological inhibitor of autophagy which blocks the fusion of autophagosomes with lysosomes and impairs the degradation of LC3-II and p62. Collectively, these results suggest that hyperglycemia leads to dysfunction of autophagy in renal tubular cells and decreases autophagic clearance. HG-induced overexpression of TXNIP may contribute to the dysfunction of tubular autophagy in diabetes.


Asunto(s)
Autofagia/fisiología , Proteínas Portadoras/fisiología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Tiorredoxinas/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Línea Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/genética , Técnicas de Silenciamiento del Gen , Humanos , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/metabolismo , Estrés Oxidativo , Fagosomas/metabolismo , Fagosomas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína Sequestosoma-1 , Tiorredoxinas/genética , Factor de Transcripción TFIIH , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Clin Sci (Lond) ; 127(7): 423-33, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24963668

RESUMEN

There is an urgent need to identify novel interventions for mitigating the progression of diabetic nephropathy. Diabetic nephropathy is characterized by progressive renal fibrosis, in which tubulointerstitial fibrosis has been shown to be the final common pathway of all forms of chronic progressive renal disease, including diabetic nephropathy. Therefore targeting the possible mechanisms that drive this process may provide novel therapeutics which allow the prevention and potentially retardation of the functional decline in diabetic nephropathy. Recently, the Ca2+-activated K+ channel KCa3.1 (KCa3.1) has been suggested as a potential therapeutic target for nephropathy, based on its ability to regulate Ca2+ entry into cells and modulate Ca2+-signalling processes. In the present review, we focus on the physiological role of KCa3.1 in those cells involved in the tubulointerstitial fibrosis, including proximal tubular cells, fibroblasts, inflammatory cells (T-cells and macrophages) and endothelial cells. Collectively these studies support further investigation into KCa3.1 as a therapeutic target in diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Animales , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Riñón/metabolismo , Riñón/patología , Riñón/fisiología , Ratones
11.
Nephrol Dial Transplant ; 29(2): 313-24, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24166472

RESUMEN

BACKGROUND: Fibroblast activation plays a critical role in diabetic nephropathy (DN). The Ca2+-activated K+ channel KCa3.1 mediates cellular proliferation of many cell types including fibroblasts. KCa3.1 has been reported to be a potential molecular target for pharmacological intervention in a diverse array of clinical conditions. However, the role of KCa3.1 in the activation of myofibroblasts in DN is unknown. These studies assessed the effect of KCa3.1 blockade on renal injury in experimental diabetes. METHODS: As TGF-ß1 plays a central role in the activation of fibroblasts to myofibroblasts in renal interstitial fibrosis, human primary renal interstitial fibroblasts were incubated with TGF-ß1+/- the selective inhibitor of KCa3.1, TRAM34, for 48 h. Two streptozotocin-induced diabetic mouse models were used in this study: wild-type KCa3.1+/+ and KCa3.1-/- mice, and secondly eNOS-/- mice treated with or without a selective inhibitor of KCa3.1 (TRAM34). Then, markers of fibroblast activation and fibrosis were determined. RESULTS: Blockade of KCa3.1 inhibited the upregulation of type I collagen, fibronectin, α-smooth muscle actin, vimentin and fibroblast-specific protein-1 in renal fibroblasts exposed to TGF-ß1 and in kidneys from diabetic mice. TRAM34 reduced TGF-ß1-induced phosphorylation of Smad2/3 and ERK1/2 but not P38 and JNK MAPK in interstitial fibroblasts. CONCLUSIONS: These results suggest that blockade of KCa3.1 attenuates diabetic renal interstitial fibrogenesis through inhibiting activation of fibroblasts and phosphorylation of Smad2/3 and ERK1/2. Therefore, therapeutic interventions to prevent or ameliorate DN through targeted inhibition of KCa3.1 deserve further consideration.


Asunto(s)
Nefropatías Diabéticas/genética , Regulación de la Expresión Génica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Corteza Renal/patología , ARN/genética , Animales , Biopsia , Western Blotting , Proliferación Celular , Células Cultivadas , Diabetes Mellitus Experimental , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Humanos , Inmunohistoquímica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Corteza Renal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Pirazoles/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
12.
Sci Rep ; 14(1): 5807, 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38461163

RESUMEN

To improve the efficiency of frozen soil excavation, the new shaft tunneling machine was developed. The new shaft tunneling machine exerts pressure on the frozen soil through the cutter under the joint action of its own gravity, the drum rotational force and the inertia force, and the frozen soil is damaged. By unique way of breaking frozen soil to improve the efficiency of frozen soil excavation, the drum rotation speed is one of the factors affecting the performance of frozen soil excavation. This article applies SolidWorks software to establish the model of cutter breaking frozen soil, takes advantage of Hyper Mesh finite element software coupled with LS-DYNA solver to acquire the regular pattern of change in the force change, frozen soil stress-strain and specific energy of cutter crushing frozen soil, etc., which analyzes the destruction of frozen soil when the drum of the new shaft tunneling machine is rotating at the speed of 25-40 rpm. Combine with field test to investigate the mechanism of cutter breaking frozen soil under the optimal drum rotation speed. The investigation results demonstrate that: when frozen soil's self-bearing capacity is lower than the force of cutter, it breaks up and detaches from the soil body, and frozen soil undergoes tensile, compressive and shear damages. For this research, it is instructive for practical engineering.

13.
Am J Physiol Renal Physiol ; 304(10): F1266-73, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23408168

RESUMEN

MicroRNAs (miRNAs) comprise of a novel class of endogenous small noncoding RNAs that frequently downregulate the expression of target genes. Recent reports suggest that miRNA-200b prevents epithelial-to-mesenchymal transition (EMT) in cancer cells by targeting the E-box binding transcription factors Zinc finger E-box-binding homeobox 1 (ZEB1) and Zinc finger E-box-binding homeobox 2 (ZEB2). About 35% of active fibroblasts are derived from EMT which is central to the development of progressive renal fibrosis. Hence, this study was designed to assess the effect of miRNA-200b on transforming growth factor-ß (TGF-ß1)-induced fibrotic responses in renal tubular cells. Morphologically, human kidney-2 cells transfected with miRNA-200b retained their epithelial cell characteristics when exposed to TGF-ß1. miRNA-200b significantly increased E-cadherin (P < 0.001) and reduced fibronectin mRNA and protein expression (both P < 0.01) independent of phospho-Smad2/3 and phospho-p38 and p42/44 signaling. Increased E-cadherin expression was associated with decreased expression of ZEB1 and ZEB2 and repression of fibronectin was mediated through direct targeting of the fibronectin mRNA, demonstrated using pMIR luciferase reporter assay and site-directed mutagenesis. These results suggest that miRNA-200b suppresses TGF-ß1-induced EMT via inhibition of ZEB1 and ZEB2 and the extracellular matrix protein fibronectin by directing targeting of its 3'UTR mRNA, independent of pathways directly involved in TGF-ß1 signaling.


Asunto(s)
Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Fibronectinas/metabolismo , Túbulos Renales Proximales/metabolismo , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibrosis , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , MicroARNs/genética , Fosforilación/efectos de los fármacos , Proteína Smad2/genética , Proteína Smad2/metabolismo , Transfección
14.
J Cutan Pathol ; 40(7): 661-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23506121

RESUMEN

The presence of non-cutaneous vascular lesions in the syndrome of multiple enchondromas and subcutaneous hemangiomas, also named Maffucci syndrome, is exceedingly rare. Until now, non-cutaneous vascular lesions have been described in nine patients, while only three cases were present in the oral cavity; they were found in the tongue in two patients and in the lower lip in one patient. Herein, we report the second case of vascular lesions localized in the mucosa of lower lip in a patient with Maffucci syndrome. Histopathologic examination showed spindle cell hemangioma.


Asunto(s)
Encondromatosis/patología , Hemangioma/patología , Neoplasias de los Labios/patología , Mucosa Bucal/patología , Neoplasias de la Boca/patología , Sarcoma/metabolismo , Adulto , Encondromatosis/metabolismo , Femenino , Hemangioma/metabolismo , Humanos , Neoplasias de los Labios/metabolismo , Mucosa Bucal/metabolismo , Neoplasias de la Boca/metabolismo , Sarcoma/patología
15.
Front Cell Dev Biol ; 11: 1302578, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37854068

RESUMEN

[This corrects the article DOI: 10.3389/fcell.2023.1293109.].

16.
Nutrients ; 15(7)2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37049564

RESUMEN

To explore the mechanism by which Akkermansia muciniphila cell-free supernatant improves glucose and lipid metabolisms in Caenorhabditis elegans, the present study used different dilution concentrations of Akkermansia muciniphila cell-free supernatant as an intervention for with Caenorhabditis elegans under a high-glucose diet. The changes in lifespan, exercise ability, level of free radicals, and characteristic indexes of glucose and lipid metabolisms were studied. Furthermore, the expression of key genes of glucose and lipid metabolisms was detected by qRT-PCR. The results showed that A. muciniphila cell-free supernatant significantly improved the movement ability, prolonged the lifespan, reduced the level of ROS, and alleviated oxidative damage in Caenorhabditis elegans. A. muciniphila cell-free supernatant supported resistance to increases in glucose and triglyceride induced by a high-glucose diet and downregulated the expression of key genes of glucose metabolism, such as gsy-1, pygl-1, pfk-1.1, and pyk-1, while upregulating the expression of key genes of lipid metabolism, such as acs-2, cpt-4, sbp-1, and tph-1, as well as down-regulating the expression of the fat-7 gene to inhibit fatty acid biosynthesis. These findings indicated that A. muciniphila cell-free supernatant, as a postbiotic, has the potential to prevent obesity and improve glucose metabolism disorders and other diseases.


Asunto(s)
Glucosa , Metabolismo de los Lípidos , Animales , Glucosa/metabolismo , Caenorhabditis elegans/metabolismo , Verrucomicrobia , Lípidos
17.
Materials (Basel) ; 15(8)2022 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-35454481

RESUMEN

The orientation, distribution, and contact point density of BF (basalt fiber) in the concrete matrix play significant roles in the mechanical properties of BF concrete, but represent a weak point in current research. It is meaningful to study the morphological characteristics of BF in concrete. In this study, the transparent model test and joint blocking method were innovatively adopted to investigate the correlation of dosage with the BF morphological parameters and concrete mechanical properties. A focus on a BF dosage of 0-7.5 kg/m3 and the contribution index of fibers Cf was defined. Furthermore, NMR and CT techniques were used to observe the changes in the microstructure of BF concrete. The experimental results show that the BF contribution index Cf reaches the largest value when the BF content is around 3 kg/m3, approximately 2.7; in this case, the mechanical properties of BF concrete were also optimal, and the Cf was only 2.34 when the BF content was 7.5 kg/m3. NMR and CT test results show that there is a strong correlation between the BF morphological parameters and the distribution of pore structure in the concrete matrix. The overlapping contact of BF clusters led to the penetration of pores, which led the macro-pore proportion to increase dramatically. The increase in the macro-pore proportion is the main reason for the deterioration in concrete performance. In addition, these macro-pores may have adverse effects on the chloride ion permeability of BF concrete.

18.
Front Med (Lausanne) ; 9: 816656, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35386912

RESUMEN

Chronic kidney disease (CKD) is rising in global prevalence and has become a worldwide public health problem, with adverse outcomes of kidney failure, cardiovascular disease, and premature death. However, current treatments are limited to slowing rather than reversing disease progression or restoring functional nephrons. Hence, innovative strategies aimed at kidney tissue recovery hold promise for CKD therapy. Mesenchymal stem cells (MSCs) are commonly used for regenerative therapy due to their potential for proliferation, differentiation, and immunomodulation. Accumulating evidence suggests that the therapeutic effects of MSCs are largely mediated by paracrine secretion of extracellular vesicles (EVs), predominantly exosomes. MSC-derived exosomes (MSC-Exos) replicate the functions of their originator MSCs via delivery of various genetic and protein cargos to target cells. More recently, MSC-Exos have also been utilized as natural carriers for targeted drug delivery. Therapeutics can be effectively incorporated into exosomes and then delivered to diseased tissue. Thus, MSC-Exos have emerged as a promising cell-free therapy in CKD. In this paper, we describe the characteristics of MSC-Exos and summarize their therapeutic efficacy in preclinical animal models of CKD. We also discuss the potential challenges and strategies in the use of MSC-Exos-based therapies for CKD in the future.

19.
Nutrients ; 14(12)2022 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-35745257

RESUMEN

Faecal microbiota transplantation (FMT) has attracted increasing attention as an intervention in many clinical conditions, including autoimmune, enteroendocrine, gastroenterological, and neurological diseases. For years, FMT has been an effective second-line treatment for Clostridium difficile infection (CDI) with beneficial outcomes. FMT is also promising in improving bowel diseases, such as ulcerative colitis (UC). Pre-clinical and clinical studies suggest that this microbiota-based intervention may influence the development and progression of chronic kidney disease (CKD) via modifying a dysregulated gut-kidney axis. Despite the high morbidity and mortality due to CKD, there are limited options for treatment until end-stage kidney disease occurs, which results in death, dialysis, or kidney transplantation. This imposes a significant financial and health burden on the individual, their families and careers, and the health system. Recent studies have suggested that strategies to reverse gut dysbiosis using FMT are a promising therapy in CKD. This review summarises the preclinical and clinical evidence and postulates the potential therapeutic effect of FMT in the management of CKD.


Asunto(s)
Infecciones por Clostridium , Colitis Ulcerosa , Insuficiencia Renal Crónica , Colitis Ulcerosa/terapia , Trasplante de Microbiota Fecal/métodos , Heces , Humanos , Diálisis Renal , Insuficiencia Renal Crónica/terapia , Resultado del Tratamiento
20.
JCI Insight ; 7(4)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35015734

RESUMEN

The G protein-coupled CXC chemokine receptor 4 (CXCR4) is a candidate therapeutic target for tissue fibrosis. A fully human single-domain antibody-like scaffold i-body AD-114-PA600 (AD-114) with specific high binding affinity to CXCR4 has been developed. To define its renoprotective role, AD-114 was administrated in a mouse model of renal fibrosis induced by folic acid (FA). Increased extracellular matrix (ECM) accumulation, macrophage infiltration, inflammatory response, TGF-ß1 expression, and fibroblast activation were observed in kidneys of mice with FA-induced nephropathy. These markers were normalized or partially reversed by AD-114 treatment. In vitro studies demonstrated AD-114 blocked TGF-ß1-induced upregulated expression of ECM, matrix metalloproteinase-2, and downstream p38 mitogen-activated protein kinase (p38 MAPK) and PI3K/AKT/mTOR signaling pathways in a renal proximal tubular cell line. Additionally, these renoprotective effects were validated in a second model of unilateral ureteral obstruction using a second generation of AD-114 (Fc-fused AD-114, also named AD-214). Collectively, these results suggest a renoprotective role of AD-114 as it inhibited the chemotactic function of CXCR4 as well as blocked CXCR4 downstream p38 MAPK and PI3K/AKT/mTOR signaling, which establish a therapeutic strategy for AD-114 targeting CXCR4 to limit renal fibrosis.


Asunto(s)
Regulación de la Expresión Génica , Enfermedades Renales/genética , Riñón/patología , Receptores CXCR4/genética , Regulación hacia Arriba , Animales , Línea Celular , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Riñón/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores CXCR4/biosíntesis , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA