Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neural Netw ; 178: 106409, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38823069

RESUMEN

Multi-center disease diagnosis aims to build a global model for all involved medical centers. Due to privacy concerns, it is infeasible to collect data from multiple centers for training (i.e., centralized learning). Federated Learning (FL) is a decentralized framework that enables multiple clients (e.g., medical centers) to collaboratively train a global model while retaining patient data locally for privacy. However, in practice, the data across medical centers are not independently and identically distributed (Non-IID), causing two challenging issues: (1) catastrophic forgetting at clients, i.e., the local model at clients will forget the knowledge received from the global model after local training, causing reduced performance; and (2) invalid aggregation at the server, i.e., the global model at the server may not be favorable to some clients after model aggregation, resulting in a slow convergence rate. To mitigate these issues, an innovative Federated learning using Model Projection (FedMoP) is proposed, which guarantees: (1) the loss of local model on global data does not increase after local training without accessing the global data so that the performance will not be degenerated; and (2) the loss of global model on local data does not increase after aggregation without accessing local data so that convergence rate can be improved. Extensive experimental results show that our FedMoP outperforms state-of-the-art FL methods in terms of accuracy, convergence rate and communication cost. In particular, our FedMoP also achieves comparable or even higher accuracy than centralized learning. Thus, our FedMoP can ensure privacy protection while outperforming centralized learning in accuracy and communication cost.

2.
Front Immunol ; 13: 1015142, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36405700

RESUMEN

Diabetic nephropathy (DN) is the most common chronic kidney disease. Accumulation of glucose and metabolites activates resident macrophages in kidneys. Resident macrophages play diverse roles on diabetic kidney injuries by releasing cytokines/chemokines, recruiting peripheral monocytes/macrophages, enhancing renal cell injuries (podocytes, mesangial cells, endothelial cells and tubular epithelial cells), and macrophage-myofibroblast transition. The differentiation and cross-talks of macrophages ultimately result renal inflammation and fibrosis in DN. Emerging evidence shows that targeting macrophages by suppressing macrophage activation/transition, and macrophages-cell interactions may be a promising approach to attenuate DN. In the review, we summarized the diverse roles of macrophages and the cross-talks to other cells in DN, and highlighted the therapeutic potentials by targeting macrophages.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Ratones , Animales , Nefropatías Diabéticas/metabolismo , Células Endoteliales/metabolismo , Ratones Endogámicos C57BL , Macrófagos/metabolismo , Riñón/metabolismo , Diabetes Mellitus/metabolismo
3.
Front Genet ; 12: 736156, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630525

RESUMEN

Kidney renal clear cell carcinoma (KIRC) is the most common malignant kidney tumor as its characterization of highly metastatic potential. Patients with KIRC are associated with poor clinical outcomes with limited treatment options. Up to date, the underlying molecular mechanisms of KIRC pathogenesis and progression are still poorly understood. Instead, particular features of Cancer-Associated Fibroblasts (CAFs) are highly associated with adverse outcomes of patients with KIRC, while the precise regulatory mechanisms at the epigenetic level of KIRC in governing CAFs remain poorly defined. Therefore, explore the correlations between epigenetic regulation and CAFs infiltration may help us better understand the molecular mechanisms behind KIRC progression, which may improve clinical outcomes and patients quality of life. In the present study, we identified a set of clinically relevant CAFs-related methylation-driven genes, NAT8, TINAG, and SLC17A1 in KIRC. Our comprehensive in silico analysis revealed that the expression levels of NAT8, TINAG, and SLC17A1 are highly associated with outcomes of patients with KIRC. Meanwhile, their methylation levels are highly correlates with the severity of KIRC. We suggest that the biomarkers might contribute to CAFs infiltration in KIRC. Taken together, our study provides a set of promising biomarkers which could predict the progression and prognosis of KIRC. Our findings could have potential prognosis and therapeutic significance in the progression of KIRC.

4.
Int J Biol Sci ; 17(14): 3911-3922, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671208

RESUMEN

Introduction and Aims: Elevated plasma levels of C-reactive protein (CRP) are closely associated with progressive renal injury in patients with chronic kidney disease (CKD). Here, we tested a hypothesis that CRP may promote renal fibrosis and inflammation via a TGF-ß/Smad3-dependent mechanism. Methods: Role and mechanisms of TGF-ß/Smad3 in CRP-induced renal fibrosis and inflammation were examined in a mouse model of unilateral ureteral obstruction (UUO) induced in CRP Tg/Smad3 KO mice and in a rat tubular epithelial cell line in which Smad3 gene is stably knocked down (S3KD-NRK52E). Results: We found that mice overexpressing the human CRP gene were largely promoted renal inflammation and fibrosis as evidenced by increasing IL-1ß, TNF-α, MCP-1 expression, F4/80+ macrophages infiltration, and marked accumulation of α-smooth muscle actin (α-SMA), collagen I and fibronectin in the UUO kidney, which were blunted when Smad3 gene was deleted in CRPtg-Smad3KO. Mechanistically, we found that the protection of renal inflammation and fibrosis in the UUO kidney of CRPtg-Smad3KO mice was associated with the inactivation of CD32-NF-κB and TGF-ß/Smad3 signaling. Conclusion: In conclusion, Smad3 deficiency protects against CRP-mediated renal inflammation and fibrosis in the UUO kidney by inactivating CD32-NF-κB and TGF-ß/Smad3 signaling.


Asunto(s)
Proteína C-Reactiva/metabolismo , Fibrosis/prevención & control , Eliminación de Gen , Inflamación/prevención & control , Enfermedades Renales/prevención & control , Proteína smad3/genética , Obstrucción Ureteral/prevención & control , Animales , Línea Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Noqueados , Ratas
5.
Int J Biol Sci ; 17(13): 3583-3594, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512167

RESUMEN

TGF-ß1 has long been considered as a key mediator in diabetic kidney disease (DKD) but anti-TGF-ß1 treatment fails clinically, suggesting a diverse role for TGF-ß1 in DKD. In the present study, we examined a novel hypothesis that latent TGF-ß1 may be protective in DKD mice overexpressing human latent TGF-ß1. Streptozotocin-induced Type 1 diabetes was induced in latent TGF-ß1 transgenic (Tg) and wild-type (WT) mice. Surprisingly, compared to WT diabetic mice, mice overexpressing latent TGF-ß1 were protected from the development of DKD as demonstrated by lowing microalbuminuria and inhibiting renal fibrosis and inflammation, although blood glucose levels were not altered. Mechanistically, the renal protective effects of latent TGF-ß1 on DKD were associated with inactivation of both TGF-ß/Smad and nuclear factor-κB (NF-κB) signaling pathways. These protective effects were associated with the prevention of renal Smad7 from the Arkadia-induced ubiquitin proteasomal degradation in the diabetic kidney, suggesting protection of renal Smad7 from Arkadia-mediated degradation may be a key mechanism through which latent TGF-ß1 inhibits DKD. This was further confirmed in vitro in mesangial cells that knockdown of Arkadia failed but overexpression of Arkadia reversed the protective effects of latent TGF-ß1 on high glucose-treated mesangial cells. Latent TGF-ß1 may protect kidneys from TGF-ß1/Smad3-mediated renal fibrosis and NF-κB-driven renal inflammation in diabetes through inhibiting Arkadia-mediated Smad7 ubiquitin degradation.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Proteínas de Unión a TGF-beta Latente/metabolismo , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/complicaciones , Nefropatías Diabéticas/genética , Proteínas de Unión a TGF-beta Latente/genética , Masculino , Ratones Endogámicos ICR , Ratones Transgénicos , FN-kappa B/metabolismo , Proteína smad7/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
6.
Exp Cell Res ; 395(1): 112170, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32682783

RESUMEN

Colorectal cancer is the second leading cause of cancer mortality worldwide with poor prognosis and high recurrence. Aberrant Wnt/ß-catenin signaling promotes oncogenesis by transcriptional activation of c-Myc and its downstream signals. EDAR is characterized as an important effector of canonical Wnt signaling in developing skin appendages, but the interplay between EDAR and Wnt signaling in tumorigenesis and progression remains to be elucidated. In this study, we revealed that EDAR expression is prevalently elevated in colorectal cancer tissues compared with normal tissues. Further analysis suggests there is a strict correlation between EDAR expression and colorectal cancer progression. EDAR silencing by shRNA in colorectal cancer cells showed proliferative suppression via retarding cell cycle at G1 phase. Xenograft mice transplanted with shEDAR-transduced tumor cells significantly alleviated tumor burden in comparison with control mice. Furthermore, downregulation of EDAR was accompanied by reduction of ß-catenin, c-Myc and other G1 cell cycle regulators, while ß-catenin agonist restored the expression of these proteins and overrode the proliferative block induced by EDAR knockdown. These findings indicate that EDAR functions as a component of Wnt/ß-catenin signaling pathway, and is a potential modulator in colorectal carcinogenesis.


Asunto(s)
Proliferación Celular/fisiología , Neoplasias del Colon , Neoplasias Colorrectales/patología , Recurrencia Local de Neoplasia/metabolismo , Receptores de la Ectodisplasina/metabolismo , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Recurrencia Local de Neoplasia/genética , Receptores de la Ectodisplasina/genética , Vía de Señalización Wnt/genética
7.
Metabolism ; 103: 154013, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31734275

RESUMEN

BACKGROUND: Transforming growth factor (TGF)-ß/Smad3 signaling is highly activated in kidneys of patients with type 2 diabetic nephropathy (T2DN), however, the precise role of Smad3 in the pathogenesis of diabetic nephropathy remains unclear. METHODS: Smad3 knockout (KO)-db/db mice were generated by intercrossing of male and female double-heterozygous Smad3+/- db/m mice. Renal functions including urinary albumin excretion and serum creatinine were determined. Renal histological injury including renal fibrosis and inflammation were examined by periodic acid Schiff (PAS), periodic acid-silver methenamine (PASM), and immunohistochemistry (IHC) staining. RESULTS: Smad3 knockout (KO)-db/db mice were protected from the development of diabetic kidney injury, characterized by the normal levels of urinary albumin excretion and serum creatinine without any evidence for renal fibrosis and inflammation. In contrast, Smad3 wild-type (WT) db/db and Smad3+/- db/db mice developed progressively decline in renal function over the 12 to 32-week time course, including increased microalbuminuria and elevated levels of serum creatinine. Pathologically, Smad3 WT db/db and Smad3+/- db/db mice exhibited a marked deposition of collagen-I (colI), collagen-IV(col-IV), and an increased infiltration of F4/80+ macrophages in kidney. Mechanistically, Smad3 deficiency decreased the lncRNA Erbb4-IR transcription, while increased miR-29b transcription and therefore protected the kidney from progressive renal injury in db/db mice. CONCLUSION: Results from this study imply that Smad3 may represent as a novel and effective therapeutic target for T2DN.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/prevención & control , Riñón/patología , Proteína smad3/genética , Albuminuria/complicaciones , Albuminuria/genética , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Femenino , Fibrosis/genética , Fibrosis/prevención & control , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
8.
Int J Med Sci ; 16(8): 1078-1088, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31523169

RESUMEN

Yunnan Baiyao (YB) as a kind of famous Chinese herbal medicine, possessed hemostatic, invigorating the circulation of blood, and anti-inflammatory effects. Identifying strategies to protect patients at risk for hospital-acquired pressure ulcers (HAPU) is essential. Herein, our results showed that YB treatment can effectively reduce the acne wound area and improve efficacy in a comparative study of 60 cases HAPU patients with S. aureus positive of acne wound pathogens. Furthermore, YB inhibited HIa expression and suppressed accessory gene regulator (agr) system controlled by regulatory RNA II and RNA III molecule using pALC1740, pALC1742 and pALC1743 S. aureus strain linked to gfpuvr reporter gene. Moreover, YB downregulated cao mRNA expression and inhibited coagulase activity by RT-PCR, slide and tube coagulase test. Additionally, YB downregulated seb, sec, sed, and tsst-1 mRNA expression to suppress enterotoxin and tsst-1 secretion and adhesion function related genes sarA, icaA, and cidA mRNA expression. Taken together, the data suggest that YB may reduce HAPU via suppressing virulence gene expression and biofilm formation of S. aureus.


Asunto(s)
Antibacterianos/farmacología , Medicamentos Herbarios Chinos/farmacología , Úlcera por Presión/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Anciano , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Biopelículas/efectos de los fármacos , Femenino , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Proteínas Hemolisinas/genética , Humanos , Enfermedad Iatrogénica , Masculino , Persona de Mediana Edad , Úlcera por Presión/microbiología , Conejos , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Transactivadores/genética , Resultado del Tratamiento , Virulencia/genética
9.
J Cell Mol Med ; 23(8): 5576-5587, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31211499

RESUMEN

Obstructive nephropathy is the end result of a variety of diseases that block drainage from the kidney(s). Transforming growth factor-ß1 (TGF-ß1)/Smad3-driven renal fibrosis is the common pathogenesis of obstructive nephropathy. In this study, we identified petchiether A (petA), a novel small-molecule meroterpenoid from Ganoderma, as a potential inhibitor of TGF-ß1-induced Smad3 phosphorylation. The obstructive nephropathy was induced by unilateral ureteral obstruction (UUO) in mice. Mice received an intraperitoneal injection of petA/vehicle before and after UUO or sham operation. An in vivo study revealed that petA protected against renal inflammation and fibrosis by reducing the infiltration of macrophages, inhibiting the expression of proinflammatory cytokines (interleukin-1ß and tumour necrosis factor-α) and reducing extracellular matrix deposition (α-smooth muscle actin, collagen I and fibronectin) in the obstructed kidney of UUO mice; these changes were associated with suppression of Smad3 and NF-κB p65 phosphorylation. Petchiether A inhibited Smad3 phosphorylation in vitro and down-regulated the expression of the fibrotic marker collagen I in TGF-ß1-treated renal epithelial cells. Further, we found that petA dose-dependently suppressed Smad3-responsive promoter activity, indicating that petA inhibits gene expression downstream of the TGF-ß/Smad3 signalling pathway. In conclusion, our findings suggest that petA protects against renal inflammation and fibrosis by selectively inhibiting TGF-ß/Smad3 signalling.


Asunto(s)
Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Terpenos/uso terapéutico , Factor de Crecimiento Transformador beta/metabolismo , Actinas/metabolismo , Animales , Línea Celular , Colágeno/metabolismo , Fibronectinas/metabolismo , Fibrosis , Humanos , Inflamación/patología , Riñón/lesiones , Riñón/patología , Enfermedades Renales/patología , Masculino , Ratones Endogámicos C57BL , Fosforilación , Terpenos/química , Terpenos/farmacología , Terpenos/toxicidad , Obstrucción Ureteral/tratamiento farmacológico , Obstrucción Ureteral/patología
10.
Mol Ther ; 26(1): 148-161, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29102563

RESUMEN

Transforming growth factor ß (TGF-ß)/Smad3 signaling plays a role in tissue fibrosis. We report here that Erbb4-IR is a novel long non-coding RNA (lncRNA) responsible for TGF-ß/Smad3-mediated renal fibrosis and is a specific therapeutic target for chronic kidney disease. Erbb4-IR was induced by TGF-ß1 via a Smad3-dependent mechanism and was highly upregulated in the fibrotic kidney of mouse unilateral ureteral obstructive nephropathy (UUO). Silencing Erbb4-IR blocked TGF-ß1-induced collagen I and alpha-smooth muscle actin (α-SMA) expressions in vitro and effectively attenuated renal fibrosis in the UUO kidney by blocking TGF-ß/Smad3 signaling. Mechanistic studies revealed that Smad7, a downstream negative regulator of TGF-ß/Smad signaling, is a target gene of Erbb4-IR because a binding site of Erbb4-IR was found on the 3' UTR of Smad7 gene. Mutation of this binding site prevented the suppressive effect of Erbb4-IR on the Smad7 reporter activity; in contrast, overexpression of Erbb4-IR largely inhibited Smad7 but increased collagen I and α-SMA transcriptions. Thus, kidney-specific silencing of Erbb4-IR upregulated renal Smad7 and thus blocked TGF-ß/Smad3-mediated renal fibrosis in vivo and in vitro. In conclusion, the present study identified that Erbb4-IR is a novel lncRNA responsible for TGF-ß/Smad3-mediated renal fibrosis by downregulating Smad7. Targeting Erbb4-IR may represent a precise therapeutic strategy for progressive renal fibrosis.


Asunto(s)
Enfermedades Renales/genética , Enfermedades Renales/metabolismo , ARN Largo no Codificante/genética , Receptor ErbB-4/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Biopsia , Línea Celular , Fibrosis , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Enfermedades Renales/patología , Ratones , Transcripción Genética
11.
Sci Rep ; 6: 27745, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27283954

RESUMEN

This study aimed to evaluate the validation of the leptin receptor-deficient mice model for secondary osteoporosis associated with type 2 diabetes mellitus (T2DM) at bone micro-architectural level. Thirty three 36-week old male mice were divided into four groups: normal control (db/m) (n = 7), leptin receptor-deficient T2DM (db/db) (n = 8), human C-reactive protein (CRP) transgenic normal control (crp/db/m) (n = 7), and human CRP transgenic T2DM (crp/db/db) (n = 11). Lumber vertebrae (L5) and bilateral lower limbs were scanned by micro-CT to analyze trabecular and cortical bone quality. Right femora were used for three-point bending to analyze the mechanical properties. Trabecular bone quality at L5 was better in db/db or crp/db/db group in terms of bone mineral density (BMD), bone volume fraction, connectivity density, trabecular number and separation (all p < 0.05). However the indices measured at proximal tibia showed comparable trabecular BMD and microarchitecture among the four groups. Femur length in crp/db/db group was significantly shorter than db/m group (p < 0.05) and cortices were thinner in db/db and crp/db/db groups (p > 0.05). Maximum loading and energy yield in mechanical test were similar among groups while the elastic modulus in db/db and crp/db/db significantly lower than db/m. The leptin-receptor mice is not a proper model for secondary osteoporosis associated with T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/patología , Osteoporosis/complicaciones , Osteoporosis/patología , Receptores de Leptina/deficiencia , Animales , Fenómenos Biomecánicos , Peso Corporal , Densidad Ósea , Huesos/diagnóstico por imagen , Huesos/patología , Huesos/fisiopatología , Hueso Esponjoso/patología , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Imagenología Tridimensional , Masculino , Ratones , Ratones Noqueados , Osteoporosis/diagnóstico por imagen , Osteoporosis/fisiopatología , Receptores de Leptina/metabolismo , Reproducibilidad de los Resultados , Microtomografía por Rayos X
12.
Sci Rep ; 6: 26740, 2016 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-27221338

RESUMEN

C-reactive protein (CRP) is associated with progressive diabetic nephropathy in patients with type-2 diabetes (T2DN). However, role of CRP in T2DN remains unclear. We report here that CRP is pathogenic in T2DN in db/db mice that express human CRP (CRPtg-db/db). Compared to the littermate db/db mice, CRPtg-db/db developed more severe T2DN, showing higher levels of fasting blood glucose and microalbuminuria and more progressive renal inflammation and fibrosis. Enhanced T2DN in CRPtg-db/db mice were associated with over-activation of CRP-CD32b, NF-κB, TGF-ß/Smad3, and mTOR signaling. Further studies in vitro defined that CRP activated Smad3 directly at 15 mins via the CD32b- ERK/p38 MAP kinase crosstalk pathway and indirectly at 24 hours through a TGF-ß1-dependent mechanism. Importantly, CRP also activated mTOR signaling at 30 mins via a Smad3-dependent mechanism as Smad3 bound mTOR physically and CRP-induced mTOR signaling was abolished by a neutralizing CD32b antibody and a specific Smad3 inhibitor. Finally, we also found that CRP induced renal fibrosis through a CD32b-Smad3-mTOR pathway because blocking mTOR signaling with rapamycin inhibited CRP-induced CTGF and collagen I expression. Thus, CRP is pathogenic in T2DN. CRP may promote CD32b- NF-κB signaling to mediate renal inflammation; whereas, CRP may enhance renal fibrosis in T2DN via CD32b-Smad3-mTOR signaling.


Asunto(s)
Proteína C-Reactiva/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/metabolismo , Receptores de IgG/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proteína C-Reactiva/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Nefropatías Diabéticas/genética , Masculino , Ratones , Ratones Transgénicos , Receptores de IgG/genética , Proteína smad3/genética , Serina-Treonina Quinasas TOR/genética
13.
Mol Ther ; 22(4): 842-53, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24445937

RESUMEN

Inflammation and its consequent fibrosis are two main features of diabetic nephropathy (DN), but target therapy on these processes for DN remains yet ineffective. We report here that miR-29b is a novel therapeutic agent capable of inhibiting progressive renal inflammation and fibrosis in type 2 diabetes in db/db mice. Under diabetic conditions, miR-29b was largely downregulated in response to advanced glycation end (AGE) product, which was associated with upregulation of collagen matrix in mesangial cells via the transforming growth factor-ß (TGF-ß)/Smad3-dependent mechanism. These pathological changes were reversed by overexpressing miR-29b, but enhanced by knocking-down miR-29b. Similarly, loss of renal miR-29b was associated with progressive diabetic kidney injury, including microalbuminuria, renal fibrosis, and inflammation. Restored renal miR-29b by the ultrasound-based gene therapy was capable of attenuating diabetic kidney disease. Further studies revealed that inhibition of Sp1 expression, TGF-ß/Smad3-dependent renal fibrosis, NF-κB-driven renal inflammation, and T-bet/Th1-mediated immune response may be mechanisms associated with miR-29b treatment in db/db mice. In conclusion, miR-29b may play a protective role in diabetic kidney disease and may have therapeutic potential for diabetic kidney complication.


Asunto(s)
Diabetes Mellitus Tipo 2/terapia , Nefropatías Diabéticas/genética , Inflamación/terapia , MicroARNs/uso terapéutico , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/terapia , Modelos Animales de Enfermedad , Fibrosis/genética , Fibrosis/terapia , Terapia Genética , Humanos , Inflamación/genética , Inflamación/patología , Ratones , MicroARNs/genética , Transducción de Señal , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética
14.
Cell Biol Int ; 35(9): 953-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21323641

RESUMEN

Proteinuria is a common feature for almost all glomerular diseases and reflects the severity of the glomerular lesion. The presence of a large amount of proteins in tubular fluid, however, may also contribute to the development of RIF (renal interstitial fibrosis). Endocytosis of albumin in proximal tubular cells triggers PKC (protein kinase C)-dependent generation of reactive oxygen species and secretion of chemokines. As a family including 12 isozymes, which PKC isozymes participate in RIF is still unclear. EMT (epithelial-mesenchymal transdifferentiation) of RTECs (renal tubular epithelial cells) plays a crucial role in the progress of RIF induced by proteinuria. In the present study, we investigated the role of classical PKC isozymes in the proteinuria-induced EMT of RTECs. Employing immunochemical staining, we found that PKC-α, -ßI and -ßII were expressed in glomerulus and in RTECs in both normal and diseased renal tissues, while PKC-γ was only expressed in podocytes in the glomerulus. Treatment of HK-2 cells with extracted urinary proteins resulted in EMT, as evidenced by morphological changes, decreased E-cadherin expression, increased α-SMA (α-smooth muscle actin) expression, as well as production of type I collagen and fibronectin. Western blot analysis of PKC isozymes in the cytosolic compared with membrane fraction revealed translocation of PKC-α and -ßI, but not PKC-ßII, in HK-2 cells undergoing EMT. Pretreatment with selective PKC-α inhibitor G-6976 or PKC-ß inhibitor significantly attenuated EMT induced by urinary proteins. In summary, the present study suggested that PKC-α and -ßI play critical roles in the EMT of RTECs in response to urinary proteins.


Asunto(s)
Células Epiteliales/citología , Transición Epitelial-Mesenquimal , Mesodermo/citología , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C/metabolismo , Transdiferenciación Celular , Células Epiteliales/metabolismo , Humanos , Isoenzimas/metabolismo , Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Mesodermo/metabolismo , Proteína Quinasa C/orina , Proteína Quinasa C beta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA