Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.305
Filtrar
1.
Signal Transduct Target Ther ; 9(1): 94, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38644381

RESUMEN

Much effort has been made to uncover the cellular heterogeneities of human hearts by single-nucleus RNA sequencing. However, the cardiac transcriptional regulation networks have not been systematically described because of the limitations in detecting transcription factors. In this study, we optimized a pipeline for isolating nuclei and conducting single-nucleus RNA sequencing targeted to detect a higher number of cell signal genes and an optimal number of transcription factors. With this unbiased protocol, we characterized the cellular composition of healthy human hearts and investigated the transcriptional regulation networks involved in determining the cellular identities and functions of the main cardiac cell subtypes. Particularly in fibroblasts, a novel regulator, PKNOX2, was identified as being associated with physiological fibroblast activation in healthy hearts. To validate the roles of these transcription factors in maintaining homeostasis, we used single-nucleus RNA-sequencing analysis of transplanted failing hearts focusing on fibroblast remodelling. The trajectory analysis suggested that PKNOX2 was abnormally decreased from fibroblast activation to pathological myofibroblast formation. Both gain- and loss-of-function in vitro experiments demonstrated the inhibitory role of PKNOX2 in pathological fibrosis remodelling. Moreover, fibroblast-specific overexpression and knockout of PKNOX2 in a heart failure mouse model induced by transverse aortic constriction surgery significantly improved and aggravated myocardial fibrosis, respectively. In summary, this study established a high-quality pipeline for single-nucleus RNA-sequencing analysis of heart muscle. With this optimized protocol, we described the transcriptional regulation networks of the main cardiac cell subtypes and identified PKNOX2 as a novel regulator in suppressing fibrosis and a potential therapeutic target for future translational studies.


Asunto(s)
Fibrosis , Proteínas de Homeodominio , Miocardio , Humanos , Ratones , Animales , Fibrosis/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Miocardio/patología , Miocardio/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ratones Noqueados , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/metabolismo , Modelos Animales de Enfermedad , Masculino
2.
J Transl Med ; 22(1): 161, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38365674

RESUMEN

BACKGROUND: The autophagy adapter SQSTM1/p62 is crucial for maintaining homeostasis in various organs and cells due to its protein-protein interaction domains and involvement in diverse physiological and pathological processes. Vascular endothelium cells play a unique role in vascular biology and contribute to vascular health. METHODS: Using the Cre-loxP system, we generated mice with endothelium cell-specific knockout of p62 mediated by Tek (Tek receptor tyrosine kinase)-cre to investigate the essential role of p62 in the endothelium. In vitro, we employed protein mass spectrometry and IPA to identify differentially expressed proteins upon knockdown of p62. Immunoprecipitation assays were conducted to demonstrate the interaction between p62 and FN1 or LAMC2 in human umbilical vein endothelium cells (HUVECs). Additionally, we identified the degradation pathway of FN1 and LAMC2 using the autophagy inhibitor 3-methyladenine (3-MA) or proteasome inhibitor MG132. Finally, the results of immunoprecipitation demonstrated that the interaction between p62 and LAMC2 was abolished in the PB1 truncation group of p62, while the interaction between p62 and FN1 was abolished in the UBA truncation group of p62. RESULTS: Our findings revealed that p62 Endo mice exhibited heart, lung, and kidney fibrosis compared to littermate controls, accompanied by severe cardiac dysfunction. Immunoprecipitation assays provided evidence of p62 acting as an autophagy adapter in the autophagy-lysosome pathway for FN1 and LAMC2 degradation respectively through PB1 and UBA domain with these proteins rather than proteasome system. CONCLUSIONS: Our study demonstrates that defects in p62 within endothelium cells induce multi-organ fibrosis and cardiac dysfunction in mice. Our findings indicate that FN1 and LAMC2, as markers of (EndoMT), have detrimental effects on HUVECs and elucidate the autophagy-lysosome degradation mechanism of FN1 and LAMC2.


Asunto(s)
Cardiopatías , Proteína Sequestosoma-1 , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Endotelio/metabolismo , Cardiopatías/genética , Cardiopatías/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/farmacología , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Fibrosis/genética , Fibrosis/metabolismo
3.
Nat Commun ; 15(1): 606, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38242884

RESUMEN

Hematopoietic mutations in epigenetic regulators like DNA methyltransferase 3 alpha (DNMT3A), play a pivotal role in driving clonal hematopoiesis of indeterminate potential (CHIP), and are associated with unfavorable outcomes in patients suffering from heart failure (HF). However, the precise interactions between CHIP-mutated cells and other cardiac cell types remain unknown. Here, we identify fibroblasts as potential partners in interactions with CHIP-mutated monocytes. We used combined transcriptomic data derived from peripheral blood mononuclear cells of HF patients, both with and without CHIP, and cardiac tissue. We demonstrate that inactivation of DNMT3A in macrophages intensifies interactions with cardiac fibroblasts and increases cardiac fibrosis. DNMT3A inactivation amplifies the release of heparin-binding epidermal growth factor-like growth factor, thereby facilitating activation of cardiac fibroblasts. These findings identify a potential pathway of DNMT3A CHIP-driver mutations to the initiation and progression of HF and may also provide a compelling basis for the development of innovative anti-fibrotic strategies.


Asunto(s)
ADN Metiltransferasa 3A , Insuficiencia Cardíaca , Humanos , Hematopoyesis Clonal , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A/genética , Fibroblastos , Fibrosis/genética , Fibrosis/patología , Insuficiencia Cardíaca/genética , Hematopoyesis/genética , Leucocitos Mononucleares , Mutación , Cardiopatías/genética , Cardiopatías/patología
5.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(5): 743-749, 2023 May 28.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-37539577

RESUMEN

MicroRNA (miR) is a class of highly conserved non-coding single-stranded RNA widely existing in mammals, which can negatively regulate the expression of targeting genes after transcription. As a key regulator, miR negatively regulates the expression of the targeting genes and disrupts important molecular signaling pathways, leading to the imbalance of multiple pathways such as tissue repair and inflammation involved in the fibrotic process. Among them, miR-15a/16 can participate in regulating and controlling the fibrotic process of various organs, including liver, lung, heart, kidney and other fibrotic diseases by acting on cell proliferation and transformation, extracellular matrix proteins production and degradation, inflammation and other important cell functions. It has potential diagnostic and therapeutic value. Clarifying the biological function of miR-15a/16 and its mechanism for action and therapeutic application prospects in various fibrotic lesions are of great significance for the molecular targeted treatment of fibrotic diseases.


Asunto(s)
Fibrosis , MicroARNs , Humanos , Fibrosis/genética , MicroARNs/genética , Transducción de Señal , Inflamación
6.
Int J Mol Sci ; 24(14)2023 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-37511199

RESUMEN

Silicosis is a refractory pneumoconiosis of unknown etiology that is characterized by diffuse lung fibrosis, and microRNA (miRNA) dysregulation is connected to silicosis. Emerging evidence suggests that miRNAs modulate pulmonary fibrosis through autophagy; however, its underlying molecular mechanism remains unclear. In agreement with miRNA microarray analysis, the qRT-PCR results showed that miR-29a-3p was significantly decreased in the pulmonary fibrosis model both in vitro and in vivo. Increased autophagosome was observed via transmission electron microscopy in lung epithelial cell models and lung tissue of silicosis mice. The expression of autophagy-related proteins LC3α/ß and Beclin1 were upregulated. The results from using 3-methyladenine, an autophagy inhibitor, or rapamycin, an autophagy inducer, together with TGF-ß1, indicated that autophagy attenuates fibrosis by protecting lung epithelial cells. In TGF-ß1-treated TC-1 cells, transfection with miR-29a-3p mimics activated protective autophagy and reduced alpha-smooth muscle actin and collagen I expression. miRNA TargetScan predicted, and dual-luciferase reporter experiments identified Akt3 as a direct target of miR-29a-3p. Furthermore, Akt3 expression was significantly elevated in the silicosis mouse model and TGF-ß1-treated TC-1 cells. The mammalian target of rapamycin (mTOR) is a central regulator of the autophagy process. Silencing Akt3 inhibited the transduction of the mTOR signaling pathway and activated autophagy in TGF-ß1-treated TC-1 cells. These results show that miR-29a-3p overexpression can partially reverse the fibrotic effects by activating autophagy of the pulmonary epithelial cells regulated by the Akt3/mTOR pathway. Therefore, targeting miR-29a-3p may provide a new therapeutic strategy for silica-induced pulmonary fibrosis.


Asunto(s)
MicroARNs , Fibrosis Pulmonar , Silicosis , Animales , Ratones , Autofagia/genética , Fibrosis/genética , Fibrosis/metabolismo , Mamíferos/metabolismo , MicroARNs/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Dióxido de Silicio/farmacología , Silicosis/etiología , Silicosis/genética , Silicosis/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Humanos
7.
Invest Ophthalmol Vis Sci ; 64(7): 21, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37310354

RESUMEN

Purpose: The purpose of this study was to investigate the role of lncRNA H19 in epithelial-mesenchymal transition (EMT) and its molecular mechanism in fibrotic cataracts. Methods: TGF-ß2-induced EMT was induced in human lens epithelial cell line (HLECs) and rat lens explants to mimic posterior capsular opacification (PCO) in vitro and in vivo. Anterior subcapsular cataract (ASC) was induced in C57BL/6J mice. The long noncoding RNA (lncRNA) H19 (H19) expression was detected by RT-qPCR. Whole-mount staining of lens anterior capsule was used to detect α-SMA and vimentin. Lentiviruses carrying shRNA or H19 vector were transfected in HLECs to knockdown or overexpress H19. Cell migration and proliferation were characterized by EdU, Transwell, and scratch assay. EMT level was detected by Western blotting and immunofluorescence. The rAAV2 carrying mouse H19 shRNA was injected into ASC model mouse anterior chambers as a gene therapy to determine its therapeutic potential. Results: PCO and ASC models were built successfully. We found H19 upregulation in PCO and ASC models in vivo and in vitro. Overexpression of H19 by lentivirus transfection increased cell migration, proliferation, and EMT. In addition, H19 knockdown by lentivirus suppressed cell migration, proliferation, and EMT levels in HLECs. Moreover, transfection of rAAV2 H19 shRNA alleviated fibrotic area in ASC mouse lens anterior capsules. Conclusions: Excessive H19 participates in lens fibrosis. Overexpression of H19 increases, whereas knockdown of H19 ameliorates HLECs migration, proliferation, and EMT. These results demonstrate H19 might be a potential target for fibrotic cataracts.


Asunto(s)
Catarata , ARN Largo no Codificante , Animales , Humanos , Ratones , Ratas , Catarata/genética , Catarata/patología , Células Epiteliales/metabolismo , Lentivirus , Ratones Endogámicos C57BL , ARN Largo no Codificante/genética , ARN Interferente Pequeño , Fibrosis/genética , Fibrosis/patología , Transición Epitelial-Mesenquimal/genética
8.
Sci Rep ; 13(1): 7701, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37169841

RESUMEN

Atrial fibrosis is a crucial contributor to initiation and perpetuation of atrial fibrillation (AF). This study aimed to identify a circRNA-miRNA-mRNA competitive endogenous RNA (ceRNA) regulatory network related to atrial fibrosis in AF, especially to validate hsa_circ_0000672/hsa_miR-516a-5p/TRAF6 ceRNA axis in AF preliminarily. The circRNA-miRNA-mRNA ceRNA network associated with AF fibrosis was constructed using bioinformatic tools and literature reviews. Left atrium (LA) low voltage was used to represent LA fibrosis by using LA voltage matrix mapping. Ten controls with sinus rhythm (SR), and 20 patients with persistent AF including 12 patients with LA low voltage and 8 patients with LA normal voltage were enrolled in this study. The ceRNA regulatory network associated with atrial fibrosis was successfully constructed, which included up-regulated hsa_circ_0000672 and hsa_circ_0003916, down-regulated miR-516a-5p and five up-regulated hub genes (KRAS, SMAD2, TRAF6, MAPK11 and SMURF1). In addition, according to the results of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, these hub genes were clustered in TGF-beta and MAPK signaling pathway. In the patients with persistent AF, hsa_circ_0000672 expression in peripheral blood monocytes was significantly higher than those in controls with SR by quantitative real-time polymerase chain reaction (p-value < 0.001). Furthermore, hsa_circ_0000672 expression was higher in peripheral blood monocytes of persistent AF patients with LA low voltage than those with LA normal voltage (p-value = 0.002). The dual-luciferase activity assay confirmed that hsa_circ_0000672 exerted biological functions as a sponge of miR-516a-5p to regulate expression of its target gene TRAF6. Hsa_circ_0000672 expression in peripheral blood monocytes may be associated with atrial fibrosis. The hsa_circ_0000672 may be involved in atrial fibrosis by indirectly regulating TRAF6 as a ceRNA by sponging miR-516a-5p.


Asunto(s)
Fibrilación Atrial , MicroARNs , Humanos , Fibrilación Atrial/genética , MicroARNs/genética , ARN Circular/genética , ARN Mensajero , Factor 6 Asociado a Receptor de TNF/genética , Ubiquitina-Proteína Ligasas , Fibrosis/genética , Fibrosis/metabolismo , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología
9.
Nat Genet ; 55(4): 665-678, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36959363

RESUMEN

After severe heart injury, fibroblasts are activated and proliferate excessively to form scarring, leading to decreased cardiac function and eventually heart failure. It is unknown, however, whether cardiac fibroblasts are heterogeneous with respect to their degree of activation, proliferation and function during cardiac fibrosis. Here, using dual recombinase-mediated genetic lineage tracing, we find that endocardium-derived fibroblasts preferentially proliferate and expand in response to pressure overload. Fibroblast-specific proliferation tracing revealed highly regional expansion of activated fibroblasts after injury, whose pattern mirrors that of endocardium-derived fibroblast distribution in the heart. Specific ablation of endocardium-derived fibroblasts alleviates cardiac fibrosis and reduces the decline of heart function after pressure overload injury. Mechanistically, Wnt signaling promotes activation and expansion of endocardium-derived fibroblasts during cardiac remodeling. Our study identifies endocardium-derived fibroblasts as a key fibroblast subpopulation accounting for severe cardiac fibrosis after pressure overload injury and as a potential therapeutic target against cardiac fibrosis.


Asunto(s)
Cardiopatías , Fibroblastos/metabolismo , Cardiopatías/genética , Cardiopatías/patología , Fibrosis/genética , Animales , Ratones , Envejecimiento , Proliferación Celular , Vía de Señalización Wnt , Ratones Transgénicos
10.
Ren Fail ; 45(1): 2187236, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36883360

RESUMEN

Type XXVIII collagen (COL28) is involved in cancer and lung fibrosis. COL28 polymorphisms and mutations might be involved in kidney fibrosis, but the exact role of COL28 in renal fibrosis is unknown. This study explored the function of COL28 in renal tubular cells by examining the expression of COL28 mRNA and the effects of COL28 overexpression in human tubular cells. COL28 mRNA expression and localization were observed in normal and fibrotic kidney tissues from humans and mice using real-time PCR, western blot, immunofluorescence, and immunohistochemistry. The consequences of COL28 overexpression on cell proliferation, migration, cell polarity, and epithelial-to-mesenchymal transition (EMT) induced by TGF-ß1 were examined in human tubular HK-2 cells. COL28 expression was low in human normal renal tissues, mainly observed in the renal tubular epithelial cells and especially in proximal renal tubules. COL28 protein expression in human and mouse obstructive kidney disease was higher than in normal tissues (p < 0.05) and more significant in the UUO2-Week than the UUO1-Week group. The overexpression of COL28 promoted HK-2 cell proliferation and enhanced their migration ability (all p < 0.05). TGF-ß1 (10 ng/ml) induced COL28 mRNA expression in HK-2 cells, decreased E-cadherin and increased α-SMA in the COL28-overexpression group compared with controls (p < 0.05). ZO-1 expression decreased while COL6 increased in the COL28-overexpression group compared with controls (p < 0.05). In conclusion, COL28 overexpression promotes the migration and proliferation of renal tubular epithelial cells. The EMT could also be involved. COL28 could be a therapeutic target against renal- fibrotic diseases.


Asunto(s)
Células Epiteliales , Enfermedades Renales , Animales , Humanos , Ratones , Proliferación Celular , Transición Epitelial-Mesenquimal/genética , Fibrosis/genética , Fibrosis/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Túbulos Renales/metabolismo , Túbulos Renales/patología , ARN Mensajero , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
11.
Int J Mol Sci ; 24(3)2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36768322

RESUMEN

Obesity is an epidemic condition linked to cardiovascular disease severity and mortality. Fat localization and type represent cardiovascular risk estimators. Importantly, visceral fat secretes adipokines known to promote low-grade inflammation that, in turn, modulate its secretome and cardiac metabolism. In this regard, IL-33 regulates the functions of various immune cells through ST2 binding and-following its role as an immune sensor to infection and stress-is involved in the pro-fibrotic remodeling of the myocardium. Here we further investigated the IL-33/ST2 effects on cardiac remodeling in obesity, focusing on molecular pathways linking adipose-derived IL-33 to the development of fibrosis or hypertrophy. We analyzed the Zucker Fatty rat model, and we developed in vitro models to mimic the adipose and myocardial relationship. We demonstrated a dysregulation of IL-33/ST2 signaling in both adipose and cardiac tissue, where they affected Epac proteins and myocardial gene expression, linked to pro-fibrotic signatures. In Zucker rats, pro-fibrotic effects were counteracted by ghrelin-induced IL-33 secretion, whose release influenced transcription factor expression and ST2 isoforms balance regulation. Finally, the effect of IL-33 signaling is dependent on several factors, such as cell types' origin and the balancing of ST2 isoforms. Noteworthy, it is reasonable to state that considering IL-33 to have a unique protective role should be considered over-simplistic.


Asunto(s)
Interleucina-33 , Obesidad , Receptores de Interleucina-1 , Remodelación Ventricular , Animales , Ratas , Cardiomegalia/genética , Cardiomegalia/metabolismo , Modelos Animales de Enfermedad , Fibrosis/genética , Fibrosis/metabolismo , Ghrelina/genética , Ghrelina/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/genética , Interleucina-33/metabolismo , Miocardio/metabolismo , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo , Ratas Zucker , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Remodelación Ventricular/genética , Remodelación Ventricular/fisiología
12.
Cell Mol Life Sci ; 80(2): 50, 2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36694058

RESUMEN

The transdifferentiation from cardiac fibroblasts to myofibroblasts is an important event in the initiation of cardiac fibrosis. However, the underlying mechanism is not fully understood. Circ-sh3rf3 (circular RNA SH3 domain containing Ring Finger 3) is a novel circular RNA which was induced in hypertrophied ventricles by isoproterenol hydrochloride, and our work has established that it is a potential regulator in cardiac hypertrophy, but whether circ-sh3rf3 plays a role in cardiac fibrosis remains unclear, especially in the conversion of cardiac fibroblasts into myofibroblasts. Here, we found that circ-sh3rf3 was down-regulated in isoproterenol-treated rat cardiac fibroblasts and cardiomyocytes as well as during fibroblast differentiation into myofibroblasts. We further confirmed that circ-sh3rf3 could interact with GATA-4 proteins and reduce the expression of GATA-4, which in turn abolishes GATA-4 repression of miR-29a expression and thus up-regulates miR-29a expression, thereby inhibiting fibroblast-myofibroblast differentiation and myocardial fibrosis. Our work has established a novel Circ-sh3rf3/GATA-4/miR-29a regulatory cascade in fibroblast-myofibroblast differentiation and myocardial fibrosis, which provides a new therapeutic target for myocardial fibrosis.


Asunto(s)
Cardiomiopatías , Fibroblastos , Fibrosis , Miofibroblastos , ARN Circular , Animales , Ratas , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Fibroblastos/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Miofibroblastos/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
13.
Hepatology ; 78(5): 1625-1653, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36626642

RESUMEN

Liver fibrosis is the result of sustained chronic liver injury and inflammation leading to hepatocyte cell death followed by the formation of fibrous scars, which is the hallmark of NASH and alcoholic steatohepatitis and can lead to cirrhosis, HCC, and liver failure. Although progress has been made in understanding the pathogenesis and clinical consequences of hepatic fibrosis, therapeutic strategies for this disease are limited. Preclinical studies suggest that peroxisome proliferator-activated receptor alpha plays an important role in preventing the development of liver fibrosis by activating genes involved in detoxifying lipotoxicity and toxins, transrepressing genes involved in inflammation, and inhibiting activation of hepatic stellate cells. Given the robust preclinical data, several peroxisome proliferator-activated receptor alpha agonists have been tested in clinical trials for liver fibrosis. Here, we provide an update on recent progress in understanding the mechanisms by which peroxisome proliferator-activated receptor alpha prevents fibrosis and discuss the potential of targeting PPARα for the development of antifibrotic treatments.


Asunto(s)
Cirrosis Hepática , PPAR alfa , Humanos , Carcinoma Hepatocelular/patología , Fibrosis/etiología , Fibrosis/genética , Fibrosis/metabolismo , Inflamación/metabolismo , Hígado/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/patología , Enfermedad del Hígado Graso no Alcohólico/patología , PPAR alfa/genética , PPAR alfa/metabolismo
14.
Tissue Cell ; 81: 102005, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36608640

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a common chronic and progressive lung disease. Fibulin-2 (FBLN2) is upregulated in patients with IPF; however, its exact role in IPF remains unclear. The present study aimed to investigate the role and the regulatory mechanism of FBLN2 in TGF-ß1-induced fibrogenesis using human lung fibroblast-derived MRC-5 cells. Cell transfection was performed to regulate FBLN2 expression. Reverse transcription-quantitative PCR and western blot analyses were performed to detect the expression levels of FBLN2 and vitronectin (VTN). Cell viability and migration were determined via the Cell Counting Kit-8 and wound healing assays, respectively. Immunofluorescence was performed to detect α-smooth muscle actin (α-SMA)-positive cells. The STRING database was used to predict the interaction between FBLN2 and VTN, which was verified via the protein immunoprecipitation assay. The results demonstrated that inhibition of FBLN2 notably inhibited TGF-ß1-induced proliferation and migration, as well as downregulating the protein expression levels of MMP2 and MMP9 in MRC-5 cells. In addition, inhibition of FBLN2 suppressed the expression levels of α-SMA, collagen type 1 α1 and fibronectin. FBLN2 was demonstrated to bind to VTN and negatively regulate its expression. Furthermore, overexpression of VTN partly abolished the inhibitory effects of FBLN2 knockdown on TGF-ß1-induced proliferation, migration and fibrosis, as well as the activity of focal adhesion kinase (FAK) signaling. Taken together, the results of the present study suggest that FBLN2 knockdown can attenuate TGF-ß1-induced fibrosis in MRC-5 cells by downregulating VTN expression via FAK signaling. Thus, FBLN2 may be a potential therapeutic target for IPF treatment.


Asunto(s)
Movimiento Celular , Proteínas de la Matriz Extracelular , Factor de Crecimiento Transformador beta1 , Vitronectina , Humanos , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Vitronectina/metabolismo
15.
Am J Pathol ; 193(3): 275-285, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36586478

RESUMEN

Planar cell polarity (PCP), a process of coordinated alignment of cell polarity across the tissue plane, may contribute to the repair of renal tubules after kidney injury. Intu is a key effector protein of PCP. Herein, conditional knockout (KO) mouse models that ablate Intu specifically from kidney tubules (Intu KO) were established. Intu KO mice and wild-type littermates were subjected to unilateral renal ischemia/reperfusion injury (IRI) or unilateral ureteral obstruction. Kidney repair was evaluated by histologic, biochemical, and immunohistochemical analyses. In vitro, scratch wound healing was examined in Intu-knockdown and control renal tubular cells. Ablation of Intu in renal tubules delayed kidney repair and ameliorated renal fibrosis after renal IRI. Intu KO mice had less renal fibrosis during unilateral ureteral obstruction. Mechanistically, Intu KO kidneys had less senescence but higher levels of cell proliferation and apoptosis during kidney repair after renal IRI. In vitro, Intu knockdown suppressed scratch wound healing in renal tubular cells, accompanied by the abnormality of centrosome orientation. Together, the results provide the first evidence for the involvement of PCP in tubular repair after kidney injury, shedding light on new strategies for improving kidney repair and recovery.


Asunto(s)
Lesión Renal Aguda , Polaridad Celular , Riñón , Daño por Reperfusión , Obstrucción Ureteral , Animales , Ratones , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Polaridad Celular/genética , Polaridad Celular/fisiología , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Riñón/metabolismo , Riñón/patología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/genética , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología
16.
FASEB J ; 37(1): e22699, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36520055

RESUMEN

Cardiac fibrosis is an essential pathological process in pressure overload (PO)-induced heart failure. Recently, myocyte-fibroblast communication is proven to be critical in heart failure, in which, pathological growth of cardiomyocytes (CMs) may promote fibrosis via miRNAs-containing exosomes (Exos). Peli1 regulates the activation of NF-κB and AP-1, which has been demonstrated to engage in miRNA transcription in cardiomyocytes. Therefore, we hypothesized that Peli1 in CMs regulates the activation of cardiac fibroblasts (CFs) through an exosomal miRNA-mediated paracrine mechanism, thereby promoting cardiac fibrosis. We found that CM-conditional deletion of Peli1 improved PO-induced cardiac fibrosis. Moreover, Exos from mechanical stretch (MS)-induced WT CMs (WT MS-Exos) promote activation of CFs, Peli1-/- MS-Exos reversed it. Furthermore, miRNA microarray and qPCR analysis showed that miR-494-3p was increased in WT MS-Exos while being down regulated in Peli1-/- MS-Exos. Mechanistically, Peli1 promoted miR-494-3p expression via NF-κB/AP-1 in CMs, and then miR-494-3p induced CFs activation by inhibiting PTEN and amplifying the phosphorylation of AKT, SMAD2/3, and ERK. Collectively, our study suggests that CMs Peli1 contributes to myocardial fibrosis via CMs-derived miR-494-3p-enriched exosomes under PO, and provides a potential exosomal miRNA-based therapy for cardiac fibrosis.


Asunto(s)
Comunicación Celular , Exosomas , Insuficiencia Cardíaca , Miocitos Cardíacos , Humanos , Exosomas/genética , Exosomas/metabolismo , Fibrosis/etiología , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factor de Transcripción AP-1/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Cardiopatías/etiología , Cardiopatías/genética , Cardiopatías/metabolismo , Cardiopatías/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Comunicación Celular/genética , Comunicación Celular/fisiología
17.
Int J Mol Sci ; 23(24)2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36555676

RESUMEN

MicroRNAs (miRNAs) are a class of short, endogenous, non-coding, single-stranded RNAs that can negatively regulate the post-transcriptional expression of target genes. Among them, miR-15a/16 is involved in the regulation of the occurrence and development of fibrosis in the liver, lungs, heart, kidneys, and other organs, as well as systemic fibrotic diseases, affecting important cellular functions, such as cell transformation, the synthesis and degradation of extracellular matrix, and the release of fibrotic mediators. Therefore, this article reviews the biological characteristics of miR-15a/16 and the molecular mechanisms and functions of their dysregulation in fibrotic diseases.


Asunto(s)
Fibrosis , MicroARNs , Humanos , Fibrosis/genética , MicroARNs/genética
18.
Int J Mol Sci ; 23(21)2022 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-36361979

RESUMEN

Cardiac lymphatic vessel (LyV) remodeling as a contributor to heart failure has not been extensively evaluated in metabolic syndrome (MetS). Our studies have shown structural changes in cardiac LyV in MetS that contribute to the development of edema and lead to myocardial fibrosis. Tissue macrophages may affect LyV via secretion of various substances, including noncoding RNAs. The aim of the study was to evaluate the influence of macrophages modified by miR-31-5p, a molecule that regulates fibrosis and lymphangiogenesis, on lymphatic endothelial cells (LECs) in vitro. The experiments were carried out on the RAW 264.7 macrophage cell line and primary dermal lymphatic endothelial cells. RAW 264.7 macrophages were transfected with miR-31-5p and supernatant from this culture was used for LEC stimulation. mRNA expression levels for genes associated with lymphangiogenesis and fibrosis were measured with qRT-PCR. Selected results were confirmed with ELISA or Western blotting. miR-31-5p-modified RAW 264.7 macrophages secreted increased amounts of VEGF-C and TGF-ß and a decreased amount of IGF-1. The supernatant from miR-31-5p-modified RAW 264.7 downregulated the mRNA expression for genes regulating endothelial-to-mesenchymal transition (EndoMT) and fibrosis in LECs. Our results suggest that macrophages under the influence of miR-31-5p show the potential to inhibit LEC-dependent fibrosis. However, more studies are needed to confirm this effect in vivo.


Asunto(s)
Células Endoteliales , MicroARNs , Células Endoteliales/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Macrófagos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Animales , Ratones , Células RAW 264.7
19.
Int J Mol Sci ; 23(22)2022 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-36430607

RESUMEN

In recent years, interest in non-coding RNAs as important physiological regulators has grown significantly. Their participation in the pathophysiology of cardiovascular diseases is extremely important. Circular RNA (circRNA) has been shown to be important in the development of heart failure. CircRNA is a closed circular structure of non-coding RNA fragments. They are formed in the nucleus, from where they are transported to the cytoplasm in a still unclear mechanism. They are mainly located in the cytoplasm or contained in exosomes. CircRNA expression varies according to the type of tissue. In the brain, almost 12% of genes produce circRNA, while in the heart it is only 9%. Recent studies indicate a key role of circRNA in cardiomyocyte hypertrophy, fibrosis, autophagy and apoptosis. CircRNAs act mainly by interacting with miRNAs through a "sponge effect" mechanism. The involvement of circRNA in the development of heart failure leads to the suggestion that they may be promising biomarkers and useful targets in the treatment of cardiovascular diseases. In this review, we will provide a brief introduction to circRNA and up-to-date understanding of their role in the mechanisms leading to the development of heart failure.


Asunto(s)
Insuficiencia Cardíaca , ARN Circular , Humanos , Enfermedades Cardiovasculares/genética , Fibrosis/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , ARN Circular/metabolismo
20.
Free Radic Biol Med ; 193(Pt 2): 485-498, 2022 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-36336232

RESUMEN

Oxidative stress is a crucial mechanism in the pathophysiology of lung injury/fibrosis and diaphragmatic dysfunction. Yes-associated protein 1 (YAP1) is a key oxidative stress response regulator. However, how lung injury/fibrosis and the subsequent YAP1 silencing treatment affect diaphragmatic function remains largely uncharacterized. In this study, mice models of acute lipopolysaccharide (LPS) and paraquat exposure were used to establish acute lung injury and chronic pulmonary fibrosis. AT2 and C2C12 cells were co-cultured under LPS and paraquat challenge. YAP1 was interfered with shRNA given in vivo and verteporfin administration in vitro. Pulmonary histology, contractile properties, and cross-sectional areas (CSAs) of the diaphragm and gastrocnemius were evaluated. Histological and biochemical analyses were performed for targeted biomarker determination. We found that LPS and paraquat caused significant lung injury/fibrosis and significantly reduced the diaphragmatic-specific force and CSAs compared with the control. YAP1 silencing alleviated inflammatory cell infiltration or collagen deposition in the lungs yet worsened the already impaired diaphragmatic function by increasing inflammatory cytokines (IL-6 and TNF-α), mitochondrial reactive oxidative species (ROS) emission, protein degradation (Murf-1, atrogin-1, and calpain), and decreasing antioxidant capabilities (superoxide dismutase 2 and glutathione peroxidase). No significant improvements were observed in diaphragmatic function by transient YAP1 knockdown in the gastrocnemius. In vitro, LPS- or paraquat-caused cytotoxicity in AT2 cells was mostly alleviated by verteporfin in a concentration that was 20-fold higher than that in C2C12 cells (20 and 1 µg/mL, respectively). Finally, 0.5 µg/mL of verteporfin significantly ameliorated hydrogen peroxide-induced proteolytic activity and antioxidant enzyme suppression in C2C12 cells, whereas 2 µg/mL of verteporfin deteriorated the same. Collectively, lung injury/fibrosis adversely affects the diaphragm. YAP1 inhibition alleviates lung injury/fibrosis but worsens diaphragmatic function potentially by enhancing inflammatory cytokines and ROS-mediated protein degradation. This disparity might be attributed to differences in susceptibility to YAP1 inhibition between muscles and the lungs.


Asunto(s)
Lesión Pulmonar Aguda , Fibrosis Pulmonar , Proteínas Señalizadoras YAP , Animales , Ratones , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Antioxidantes/farmacología , Antioxidantes/metabolismo , Citocinas/metabolismo , Diafragma/metabolismo , Diafragma/fisiología , Fibrosis/genética , Inflamación/genética , Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Pulmón/metabolismo , Pulmón/patología , Estrés Oxidativo/genética , Paraquat/efectos adversos , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Especies Reactivas de Oxígeno/metabolismo , Verteporfina/efectos adversos , Verteporfina/metabolismo , Proteínas Señalizadoras YAP/genética , Proteínas Señalizadoras YAP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA