Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.574
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Development ; 146(15)2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31331942

RESUMEN

Postnatal alveolar formation is the most important and the least understood phase of lung development. Alveolar pathologies are prominent in neonatal and adult lung diseases. The mechanisms of alveologenesis remain largely unknown. We inactivated Pdgfra postnatally in secondary crest myofibroblasts (SCMF), a subpopulation of lung mesenchymal cells. Lack of Pdgfra arrested alveologenesis akin to bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease. The transcriptome of mutant SCMF revealed 1808 altered genes encoding transcription factors, signaling and extracellular matrix molecules. Elastin mRNA was reduced, and its distribution was abnormal. Absence of Pdgfra disrupted expression of elastogenic genes, including members of the Lox, Fbn and Fbln families. Expression of EGF family members increased when Tgfb1 was repressed in mouse. Similar, but not identical, results were found in human BPD lung samples. In vitro, blocking PDGF signaling decreased elastogenic gene expression associated with increased Egf and decreased Tgfb family mRNAs. The effect was reversible by inhibiting EGF or activating TGFß signaling. These observations demonstrate the previously unappreciated postnatal role of PDGFA/PDGFRα in controlling elastogenic gene expression via a secondary tier of signaling networks composed of EGF and TGFß.


Asunto(s)
Familia de Proteínas EGF/metabolismo , Miofibroblastos/metabolismo , Alveolos Pulmonares/embriología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Displasia Broncopulmonar/patología , Proteínas de Unión al Calcio/biosíntesis , Diferenciación Celular/fisiología , Células Cultivadas , Elastina/genética , Proteínas de la Matriz Extracelular/biosíntesis , Fibrilina-1/biosíntesis , Humanos , Ratones , Ratones Noqueados , Proteína-Lisina 6-Oxidasa/biosíntesis , ARN Mensajero/genética , Factor de Crecimiento Transformador beta1/biosíntesis
2.
J Am Soc Nephrol ; 32(11): 2777-2794, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34716242

RESUMEN

BACKGROUND: Autoantibodies binding to podocyte antigens cause idiopathic membranous glomerulonephritis (iMGN). However, it remains elusive how autoantibodies reach the subepithelial space because the glomerular filtration barrier (GFB) is size selective and almost impermeable for antibodies. METHODS: Kidney biopsies from patients with iMGN, cell culture, zebrafish, and mouse models were used to investigate the role of nephronectin (NPNT) regulating microRNAs (miRs) for the GFB. RESULTS: Glomerular endothelial cell (GEC)-derived miR-192-5p and podocyte-derived miR-378a-3p are upregulated in urine and glomeruli of patients with iMGN, whereas glomerular NPNT is reduced. Overexpression of miR-192-5p and morpholino-mediated npnt knockdown induced edema, proteinuria, and podocyte effacement similar to podocyte-derived miR-378a-3p in zebrafish. Structural changes of the glomerular basement membrane (GBM) with increased lucidity, splitting, and lamellation, especially of the lamina rara interna, similar to ultrastructural findings seen in advanced stages of iMGN, were found. IgG-size nanoparticles accumulated in lucidity areas of the lamina rara interna and lamina densa of the GBM in npnt-knockdown zebrafish models. Loss of slit diaphragm proteins and severe structural impairment of the GBM were further confirmed in podocyte-specific Npnt knockout mice. GECs downregulate podocyte NPNT by transfer of miR-192-5p-containing exosomes in a paracrine manner. CONCLUSIONS: Podocyte NPNT is important for proper glomerular filter function and GBM structure and is regulated by GEC-derived miR-192-5p and podocyte-derived miR-378a-3p. We hypothesize that loss of NPNT in the GBM is an important part of the initial pathophysiology of iMGN and enables autoantigenicity of podocyte antigens and subepithelial immune complex deposition in iMGN.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Membrana Basal Glomerular/metabolismo , Membrana Basal Glomerular/fisiopatología , Glomerulonefritis Membranosa/genética , Glomérulos Renales/metabolismo , MicroARNs/fisiología , Animales , Complejo Antígeno-Anticuerpo/análisis , Autoantígenos/genética , Autoantígenos/inmunología , Células Cultivadas , Técnicas de Cocultivo , Exosomas/metabolismo , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/fisiología , Regulación de la Expresión Génica , Marcación de Gen , Membrana Basal Glomerular/inmunología , Membrana Basal Glomerular/ultraestructura , Glomerulonefritis Membranosa/inmunología , Glomerulonefritis Membranosa/metabolismo , Glomerulonefritis Membranosa/fisiopatología , Tiosulfato Sódico de Oro , Humanos , Nanopartículas del Metal , Ratones , MicroARNs/biosíntesis , MicroARNs/genética , MicroARNs/orina , Comunicación Paracrina , Permeabilidad , Podocitos/inmunología , Podocitos/metabolismo , Proteinuria/etiología , Transfección , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
3.
J Neurochem ; 157(6): 1745-1758, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33113163

RESUMEN

Schizophrenia is a severe psychiatric disorder with a strong hereditary component that affects approximately 1% of the world's population. The disease is most likely caused by the altered expression of a number of genes that function at the level of biological pathways or gene networks. Transcription factors (TF) are indispensable regulators of gene expression. EGR3 is a TF associated with schizophrenia. In the current study, DNA microarray and ingenuity pathway analyses (IPA) demonstrated that EGR3 regulates Reelin signaling pathway in SH-SY5Y cells. ChIP and luciferase reporter studies confirmed that EGR3 directly binds to the promoter region of RELN thereby activating RELN expression. The expression of both EGR3 and RELN was decreased during neuronal differentiation induced by retinoic acid (RA) in SH-SY5Y cells, and EGR3 over-expression reduced neurite outgrowth which could be partially reversed by the knockdown of RELN. The expression levels of EGR3 and RELN in peripheral blood of subjects with schizophrenia were found to be down-regulated (compared with healthy controls), and were positively correlated. Furthermore, data mining from public databases revealed that the expression levels of EGR3 and RELN were presented a positive correlation in post-mortem brain tissue of subjects with schizophrenia. Taken together, this study suggests that EGR3 is a novel TF of the RELN gene and regulates neurite outgrowth via the Reelin signaling pathway. Our findings contribute to the understanding of the regulatory role of EGR3 in the pathophysiology and molecular mechanisms of schizophrenia, and potentially to the development of new therapies and diagnostic biomarkers for the disorder.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Proteína 3 de la Respuesta de Crecimiento Precoz/genética , Proteínas de la Matriz Extracelular/genética , Predisposición Genética a la Enfermedad/genética , Proteínas del Tejido Nervioso/genética , Proyección Neuronal/fisiología , Esquizofrenia/genética , Serina Endopeptidasas/genética , Transducción de Señal/fisiología , Adulto , Anciano , Moléculas de Adhesión Celular Neuronal/biosíntesis , Línea Celular Tumoral , Proteína 3 de la Respuesta de Crecimiento Precoz/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , Femenino , Regulación de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad/etnología , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/biosíntesis , Proteína Reelina , Esquizofrenia/etnología , Esquizofrenia/metabolismo , Serina Endopeptidasas/biosíntesis , Adulto Joven
4.
Circ Res ; 124(9): 1323-1336, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30832557

RESUMEN

RATIONALE: Targeting inflammation has been shown to provide clinical benefit in the field of cardiovascular diseases. Although manipulating regulatory T-cell function is an important goal of immunotherapy, the molecules that mediate their suppressive activity remain largely unknown. IL (interleukin)-35, an immunosuppressive cytokine mainly produced by regulatory T cells, is a novel member of the IL-12 family and is composed of an EBI3 (Epstein-Barr virus-induced gene 3) subunit and a p35 subunit. However, the role of IL-35 in infarct healing remains elusive. OBJECTIVE: This study aimed to determine whether IL-35 signaling is involved in healing and cardiac remodeling after myocardial infarction (MI) and, if so, to elucidate the underlying molecular mechanisms. METHODS AND RESULTS: IL-35 subunits (EBI3 and p35), which are mainly expressed in regulatory T cells, were upregulated in mice after MI. After IL-35 inhibition, mice showed impaired infarct healing and aggravated cardiac remodeling, as demonstrated by a significant increase in mortality because of cardiac rupture, decreased wall thickness, and worse cardiac function compared with wild-type MI mice. IL-35 inhibition also led to decreased expression of α-SMA (α-smooth muscle actin) and collagen I/III in the hearts of mice after MI. Pharmacological inhibition of IL-35 suppressed the accumulation of Ly6Clow and major histocompatibility complex IIlow/C-C motif chemokine receptor type 2- (MHC IIlow CCR2-) macrophages in infarcted hearts. IL-35 activated transcription of CX3CR1 (C-X3-C motif chemokine receptor 1) and TGF (transforming growth factor) ß1 in macrophages by inducing GP130 signaling, via IL12Rß2 and phosphorylation of STAT1 (signal transducer and activator of transcription family) and STAT4 and subsequently promoted Ly6Clow macrophage survival and extracellular matrix deposition. Moreover, compared with control MI mice, IL-35-treated MI mice showed increased expression of α-SMA and collagen within scars, correlating with decreased left ventricular rupture rates. CONCLUSIONS: IL-35 reduces cardiac rupture, improves wound healing, and attenuates cardiac remodeling after MI by promoting reparative CX3CR1+Ly6Clow macrophage survival.


Asunto(s)
Interleucinas/fisiología , Macrófagos/fisiología , Infarto del Miocardio/fisiopatología , Cicatrización de Heridas/fisiología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/farmacología , Receptor 1 de Quimiocinas CX3C/biosíntesis , Receptor 1 de Quimiocinas CX3C/genética , Supervivencia Celular , Cicatriz/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Regulación de la Expresión Génica/fisiología , Rotura Cardíaca Posinfarto/fisiopatología , Rotura Cardíaca Posinfarto/prevención & control , Interleucinas/antagonistas & inhibidores , Interleucinas/biosíntesis , Interleucinas/genética , Ratones , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor/biosíntesis , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Miocardio/metabolismo , Receptores de Citocinas/antagonistas & inhibidores , Receptores de Citocinas/biosíntesis , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Regulación hacia Arriba , Remodelación Ventricular/fisiología
5.
J Surg Res ; 259: 261-270, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33162101

RESUMEN

BACKGROUND: Keloid is a benign fibroproliferative tumor of the skin caused by abnormal wound healing process after skin injury. Long noncoding RNAs have been reported to be involved in the development of keloid. However, the role and mechanism of nuclear enriched abundant transcript 1 (NEAT1) in keloid are still unknown. METHODS: Quantitative real-time polymerase chain reaction was performed to detect the expression of NEAT1, miR-196b-5p, and fibroblast growth factor 2 (FGF2). Western blot was conducted to measure the levels of collagen I, α-smooth muscle actin, fibronectin, and FGF2. Cell Counting Kit-8 assay and transwell assay were used to evaluate cell viability and migration, respectively. Dual-luciferase reporter assay was conducted to verify the targeting relationship between miR-196b-5p and NEAT1 or FGF2. RESULTS: NEAT1 was increased and miR-196b-5p was decreased in keloid tissues and fibroblasts. NEAT1 knockdown or miR-196b-5p overexpression suppressed cell viability, migration, and extracellular matrix (ECM) component production in keloid fibroblasts. MiR-196 b-5p was a target of NEAT1, and NEAT1 overexpression reversed the effect of miR-196b-5p on keloid fibroblast progression. Moreover, we found that miR-196b-5p directly targeted FGF2. FGF2 knockdown suppressed keloid fibroblast viability, migration, and ECM protein production. FGF2 overexpression abolished the effect of miR-196b-5p overexpression on keloid fibroblast development. CONCLUSIONS: NEAT1 silencing suppressed cell viability, migration, and ECM expression in keloid fibroblasts by regulating miR-196b-5p/FGF2 axis, indicating a promising strategy for keloid treatment.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Queloide/patología , MicroARNs/fisiología , ARN Largo no Codificante/fisiología , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Proteínas de la Matriz Extracelular/biosíntesis , Fibroblastos/fisiología , Humanos , Queloide/metabolismo
6.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34638874

RESUMEN

Platelet concentrate products are increasingly used in many medical disciplines due to their regenerative properties. As they contain a variety of chemokines, cytokines, and growth factors, they are used to support the healing of chronic or complicated wounds. To date, underlying cellular mechanisms have been insufficiently investigated. Therefore, we analyzed the influence of Platelet-Released Growth Factors (PRGF) on human dermal fibroblasts. Whole transcriptome sequencing and gene ontology (GO) enrichment analysis of PRGF-treated fibroblasts revealed an induction of several genes involved in the formation of the extracellular matrix (ECM). Real-time PCR analyses of PRGF-treated fibroblasts and skin explants confirmed the induction of ECM-related genes, in particular transforming growth factor beta-induced protein (TGFBI), fibronectin 1 (FN1), matrix metalloproteinase-9 (MMP-9), transglutaminase 2 (TGM2), fermitin family member 1 (FERMT1), collagen type I alpha 1 (COL1A1), a disintegrin and metalloproteinase 19 (ADAM19), serpin family E member 1 (SERPINE1) and lysyl oxidase-like 3 (LOXL3). The induction of these genes was time-dependent and in part influenced by the epidermal growth factor receptor (EGFR). Moreover, PRGF induced migration and proliferation of the fibroblasts. Taken together, the observed effects of PRGF on human fibroblasts may contribute to the underlying mechanisms that support the beneficial wound-healing effects of thrombocyte concentrate products.


Asunto(s)
Plaquetas/química , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas de la Matriz Extracelular/biosíntesis , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Células Cultivadas , Cadena alfa 1 del Colágeno Tipo I , Humanos , Péptidos y Proteínas de Señalización Intercelular/química
7.
Molecules ; 26(24)2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34946675

RESUMEN

(1) Background: Cosmeceuticals are topical products applied to human skin to prevent skin ageing and maintain a healthy skin appearance. Their effectiveness is closely linked to the compounds present in a final formulation. In this article, we propose a panel of in vitro tests to support the efficacy assessment of an anti-ageing cream enriched with functional compounds. (2) Methods: biocompatibility and the irritant effect were evaluated on reconstructed human epidermis (RHE) and corneal epithelium (HCE) 3D models. After a preliminary MTT assay, normal human dermal fibroblasts (NHDF) and keratinocytes (HaCaT) were used to evaluate the extracellular matrix (ECM) protein synthesis, and interleukin-6 (IL-6) and metalloproteinase-1 (MMP-1) production. (3) Results: data collected showed good biocompatibility and demonstrated the absence of the irritant effect in both 3D models. Therefore, we demonstrated a statistical increase in collagen and elastin productions in NHDF cells. In HaCaT cells, we highlighted an anti-inflammatory effect through a reduction in IL-6 levels in inflammatory stimulated conditions. Moreover, the reduction of MMP-1 production after UV-B radiation was demonstrated, showing significant photo-protection. (4) Conclusion: a multiple in vitro assays approach is proposed for the valid and practical assessment of the anti-ageing protection, anti-inflammatory and biocompatible claims that can be assigned to a cosmetic product containing functional compounds.


Asunto(s)
Cosmecéuticos/farmacología , Dermis/metabolismo , Fibroblastos/metabolismo , Queratinocitos/metabolismo , Envejecimiento de la Piel/efectos de los fármacos , Línea Celular , Proteínas de la Matriz Extracelular/biosíntesis , Humanos , Interleucina-6/biosíntesis , Metaloproteinasa 1 de la Matriz/biosíntesis
8.
Bull Exp Biol Med ; 172(1): 96-99, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34791562

RESUMEN

Human corneal stromal cells were isolated by enzymatic digestion from a new source, lenticules obtained during laser vision correction by the ReLEx SMILe method. The resulting culture was mainly presented by fibroblast-like cells with a phenotype CD90-/CD73+/CD105+/keratocan-/lumican-/ALDH1A1+ that differentiate into keratocytes in a specialized medium. The concentration of fetal calf serum-derived growth factors affects the rate of proliferation, production of erythropoietin and brain neurotrophic factor by corneal fibroblasts, and to a lesser extent, their migration activity and production of extracellular matrix components. Thus, the high functional potential of fibroblast-like cells isolated from stromal lenticles can be used to develop cell technologies in ophthalmology.


Asunto(s)
Queratocitos de la Córnea/citología , Sustancia Propia/citología , Fibroblastos/metabolismo , Células del Estroma/citología , 5'-Nucleotidasa/metabolismo , Familia de Aldehído Deshidrogenasa 1/metabolismo , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Sustancia Propia/metabolismo , Endoglina/metabolismo , Eritropoyetina/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas Ligadas a GPI/metabolismo , Humanos , Lumican/metabolismo , Proteoglicanos/metabolismo , Retinal-Deshidrogenasa/metabolismo , Células del Estroma/metabolismo , Antígenos Thy-1/metabolismo
9.
Prostate ; 80(10): 753-763, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32421868

RESUMEN

BACKGROUND: Although thrombospondins 4 (THBS4) participates in controlling the biology of prostate cancer (PCa), the mechanism underlying this regulation remains unknown. Hence, this study aims to identify the regulatory effects of THBS4 on the PCa stem cell-like properties and the potential mechanism associated with the phosphatidylinositol 3'-kinase (PI3K)/protein kinase B (Akt) pathway. METHODS: PCa stem cells were sorted and identified using flow cytometry and THBS4 expression in the identified PCa stem cells was measured using Western blot assay. THBS4 was overexpressed or silenced in PCa stem cells, following which, self-renewal, proliferation, cell cycle distribution, and apoptosis of PCa stem cells were assessed as well as tumorigenicity in vivo was evaluated. PI3K/Akt pathway inhibitor was applied to identify its involvement in the regulatory roles of THBS4 in PCa stem cells. RESULTS: THBS4 was expressed at a higher level in PCa stem cells than in PCa cells. The overexpression of THBS4 promoted the self-renewal and proliferation, curbed the apoptosis of PCa stem cells, and enhanced the in vivo tumorigenicity, which was achieved by activating the PI3K/Akt pathway. On the contrary, short-hairpin RNA-mediated silencing of THBS4 exhibited suppressive effects on those cancer stem cell (CSC)-like properties and promotive effects on their apoptosis. CONCLUSION: THBS4 silencing can impede the CSC-like properties in PCa via blockade of the PI3K/Akt pathway, which provides patients with PCa a new therapeutic target.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trombospondinas/metabolismo , Antígeno AC133/biosíntesis , Animales , Línea Celular Tumoral , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Silenciador del Gen , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Células PC-3 , Neoplasias de la Próstata/genética , Transducción de Señal , Trombospondinas/biosíntesis , Trombospondinas/deficiencia , Trombospondinas/genética
10.
Biochem Biophys Res Commun ; 527(1): 283-288, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32446381

RESUMEN

Lysyl oxidase (LOX) is involved in fibrosis by catalyzing collagen cross-linking. Previous work observed that Triptolide (TPL) alleviated radiation-induced pulmonary fibrosis (RIPF), but it is unknown whether the anti-RIPF effect of TPL is related to LOX. In a mouse model of RIPF, we found that LOX persistently increased in RIPF which was significantly lowered by TPL. Excessive LOX aggravated fibrotic lesions in RIPF, while LOX inhibition mitigated RIPF. Irradiation enhanced the transcription and synthesis of LOX by lung fibroblasts through IKKß/NFκB activation, and siRNA knockdown IKKß largely abolished LOX production. By interfering radiation induced IKKß activation, TPL prevented NFκB nuclear translocation and DNA binding, and potently decreased LOX synthesis. Our results demonstrate that the anti-RIPF effect of TPL is associated with reduction of LOX production which mediated by inhibition of IKKß/NFκB pathway.


Asunto(s)
Diterpenos/farmacología , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Quinasa I-kappa B/antagonistas & inhibidores , Fenantrenos/farmacología , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Fibrosis Pulmonar/tratamiento farmacológico , Traumatismos por Radiación/tratamiento farmacológico , Animales , Diterpenos/administración & dosificación , Relación Dosis-Respuesta a Droga , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/farmacología , Proteínas de la Matriz Extracelular/biosíntesis , Femenino , Quinasa I-kappa B/metabolismo , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Fenantrenos/administración & dosificación , Proteína-Lisina 6-Oxidasa/biosíntesis , Fibrosis Pulmonar/metabolismo , Traumatismos por Radiación/metabolismo , Relación Estructura-Actividad
11.
Biochem Biophys Res Commun ; 528(3): 561-566, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32505347

RESUMEN

Cardiac fibroblast (CF) differentiation plays a crucial role in cardiac fibrosis, which is a specific manifestation distinguishing pathological cardiac hypertrophy from physiological hypertrophy. The DNA-binding activity of paired box 6 (Pax6) has been shown to be oppositely regulated in physiological and pathological hypertrophy; however, it remains unclear whether Pax6 is involved in CF differentiation during cardiac fibrosis. We found that Pax6 is expressed in the heart of and CFs isolated from adult mice. Moreover, angiotensin II (Ang II) induced the downregulation of Pax6 mRNA and protein expression in fibrotic heart tissue and cardiac myofibroblasts. Pax6 knockdown in CFs promoted the expression of the myofibroblast marker α-smooth muscle actin (α-SMA) and the synthesis of the extracellular matrix (ECM) proteins collagen I and fibronectin. Furthermore, we validated the ability of Pax6 to bind to the promoter regions of Cxcl10 and Il1r2 and the intronic region of Tgfb1. Pax6 knockdown in CFs decreased CXC chemokine 10 (CXCL10) and interleukin-1 receptor 2 (IL-1R2) expression and increased transforming growth factor ß1 (TGFß1) expression, mimicking the effects of Ang II. In conclusion, Pax6 exerts an inhibitory effect on CF differentiation and ECM synthesis by transcriptionally activating the expression of the anti-fibrotic factors CXCL10 and IL-1R2 and repressing the expression of the pro-fibrotic factor TGFß1. Therefore, maintaining Pax6 expression in CFs is essential for preventing CF differentiation, and provides a new therapeutic target for cardiac fibrosis.


Asunto(s)
Diferenciación Celular/fisiología , Miocardio/citología , Miocardio/metabolismo , Factor de Transcripción PAX6/fisiología , Angiotensina II/farmacología , Animales , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Diferenciación Celular/genética , Quimiocina CXCL10/genética , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/biosíntesis , Fibroblastos/citología , Fibroblastos/metabolismo , Fibrosis , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Intrones , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción PAX6/antagonistas & inhibidores , Factor de Transcripción PAX6/genética , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Receptores Tipo II de Interleucina-1/genética , Factor de Crecimiento Transformador beta1/genética
12.
Development ; 144(24): 4563-4572, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29122839

RESUMEN

Alveologenesis, the final step of lung development, is characterized by the formation of millions of alveolar septa that constitute the vast gas-exchange surface area. The genetic network driving alveologenesis is poorly understood compared with earlier steps in lung development. FGF signaling through receptors Fgfr3 and Fgfr4 is crucial for alveologenesis, but the mechanisms through which they mediate this process remain unclear. Here we show that in Fgfr3;Fgfr4 (Fgfr3;4) global mutant mice, alveolar simplification is first observed at the onset of alveologenesis at postnatal day 3. This is preceded by disorganization of elastin, indicating defects in the extracellular matrix (ECM). Although Fgfr3 and Fgfr4 are expressed in the mesenchyme and epithelium, inactivation in the mesenchyme, but not the epithelium, recapitulated the defects. Expression analysis of components of the elastogenesis machinery revealed that Mfap5 (also known as Magp2), which encodes an elastin-microfibril bridging factor, is upregulated in Fgfr3;4 mutants. Mfap5 mutation in the Fgfr3;4 mutant background partially attenuated the alveologenesis defects. These data demonstrate that, during normal lung maturation, FGF signaling restricts expression of the elastogenic machinery in the lung mesenchyme to control orderly formation of the elastin ECM, thereby driving alveolar septa formation to increase the gas-exchange surface.


Asunto(s)
Proteínas Contráctiles/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , Organogénesis/fisiología , Alveolos Pulmonares/embriología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Proteínas Contráctiles/genética , Epitelio/metabolismo , Matriz Extracelular/patología , Proteínas de la Matriz Extracelular/genética , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Alveolos Pulmonares/citología , Factores de Empalme de ARN , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/fisiología
13.
FASEB J ; 33(11): 12200-12212, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31373848

RESUMEN

In hypertrophic scar (HS) formation, the type 2 immune response induces the alternatively activated macrophages (M2), which manipulate fibroblasts to differentiate into myofibroblasts with active biologic functions and proliferation. Myofibroblasts express α-smooth muscle actin (α-SMA) and synthesize and produce additional collagen type I and collagen type III, inducing HS formation. However, studies on the mechanism of M2 macrophage modulation are only based on the recognition of profibrotic factors such as TGF-ß1 secreted by macrophages. The influence of exosomes from M2 macrophages on scar formation is still unknown. Both M2 macrophages and myofibroblasts highly express glutaminases (GLSs). GLS is a critical enzyme in glutaminolysis and is important for M2 macrophage and fibroblast polarization. In this study, we found that in a TGF-ß1-stimulated coculture system, a long noncoding RNA (lncRNA) named lncRNA-ASLNCS5088 was enriched in M2 macrophage-derived exosomes. This lncRNA could be transferred with high efficiency to fibroblasts and acted as an endogenous sponge to adsorb microRNA-200c-3p, resulting in increased GLS and α-SMA expression. Pretreatment with GW4869, which impairs M2 macrophage exosome synthesis, ameliorated these pathologic changes in fibroblasts in vitro. Local injection in the late scar formation period with GW4869 reduced α-SMA+ fibroblasts and alleviated the fibrosis of tissue after wound healing in vivo.-Chen, J., Zhou, R., Liang, Y., Fu, X., Wang, D., Wang, C. Blockade of lncRNA-ASLNCS5088-enriched exosome generation in M2 macrophages by GW4869 dampens the effect of M2 macrophages on orchestrating fibroblast activation.


Asunto(s)
Compuestos de Anilina/farmacología , Compuestos de Bencilideno/farmacología , Cicatriz Hipertrófica/etiología , Exosomas/fisiología , Fibroblastos/fisiología , Macrófagos/fisiología , ARN Largo no Codificante/fisiología , Actinas/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , Glutaminasa/biosíntesis , Humanos , Células THP-1 , Factor de Crecimiento Transformador beta1/fisiología
14.
Mol Pharm ; 17(7): 2390-2397, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32437164

RESUMEN

Opticin is an endogenous vitreous glycoprotein that may have therapeutic potential as it has been shown that supranormal concentrations suppress preretinal neovascularization. Herein we investigated the pharmacokinetics of opticin following intravitreal injection in rabbits. To measure simultaneously concentrations of human and rabbit opticin, a selected reaction monitoring mass spectrometry assay was developed. The mean concentration of endogenous rabbit opticin in 7 uninjected eyes was measured and found to be 19.2 nM or 0.62 µg/mL. When the vitreous was separated by centrifugation into a supernatant and collagen-containing pellet, 94% of the rabbit opticin was in the supernatant. Intravitreal injection of human opticin (40 µg) into both eyes of rabbits was followed by enucleation at 5, 24, and 72 h and 7, 14, and 28 days postinjection (n = 6 at each time point) and measurement of vitreous human and rabbit opticin concentrations in the supernatant and collagen-containing pellet following centrifugation. The volume of distribution of human opticin was calculated to be 3.31 mL, and the vitreous half-life was 4.2 days. Assuming that rabbit and human opticin are cleared from rabbit vitreous at the same rate, opticin is secreted into the vitreous at a rate of 0.14 µg/day. We conclude that intravitreally injected opticin has a vitreous half-life that is similar to currently available antiangiogenic therapeutics. While opticin was first identified bound to vitreous collagen fibrils, here we demonstrate that >90% of endogenous opticin is not bound to collagen. Endogenous opticin is secreted by the nonpigmented ciliary epithelium into the rabbit vitreous at a remarkably high rate, and the turnover in vitreous is approximately 15% per day.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacocinética , Proteínas de la Matriz Extracelular/administración & dosificación , Proteínas de la Matriz Extracelular/farmacocinética , Inyecciones Intravítreas/métodos , Proteoglicanos/administración & dosificación , Proteoglicanos/farmacocinética , Inhibidores de la Angiogénesis/biosíntesis , Animales , Colágeno/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/metabolismo , Semivida , Humanos , Masculino , Espectrometría de Masas/métodos , Neovascularización Fisiológica/efectos de los fármacos , Proteoglicanos/biosíntesis , Proteoglicanos/metabolismo , Conejos , Retina/metabolismo , Cuerpo Vítreo/metabolismo
15.
Mediators Inflamm ; 2020: 9578701, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32848510

RESUMEN

Collagen triple helix repeat containing-1 (CTHRC1) has been identified as cancer-related protein. CTHRC1 expresses mainly in adventitial fibroblasts and neointimal smooth muscle cells of balloon-injured vessels and promotes cell migration and tissue repair in response to injury. CTHRC1 plays a pivotal role in some pathophysiological processes, including increasing bone mass, preventing myelination, and reversing collagen synthesis in many tumor cells. The ascended expression of CTHRC1 is related to tumorigenesis, proliferation, invasion, and metastasis in various human malignancies, including gastric cancer, pancreatic cancer, hepatocellular carcinoma, keloid, breast cancer, colorectal cancer, epithelial ovarian cancer, esophageal squamous cell carcinoma, cervical cancer, non-small-cell lung carcinoma, and melanoma. And molecules that regulate the expression of CTHRC1 include miRNAs, lncRNAs, WAIF1, and DPAGT1. Many reports have pointed that CTHRC1 could exert different effects through several signaling pathways such as TGF-ß, Wnt, integrin ß/FAK, Src/FAK, MEK/ERK, PI3K/AKT/ERK, HIF-1α, and PKC-δ/ERK signaling pathways. As a participant in tissue remodeling or immune response, CTHRC1 may promote early-stage cancer. Several recent studies have identified CTHRC1 as an effectual prognostic biomarker for predicting tumor recurrence or metastasis. It is worth noting that CTHRC1 has different cellular localization and mechanisms of action in different cells and different microenvironments. In this article, we focus on the advances in the signaling pathways mediated by CTHRC1 in tumors.


Asunto(s)
Progresión de la Enfermedad , Proteínas de la Matriz Extracelular/biosíntesis , Regulación de la Expresión Génica , Metástasis de la Neoplasia , Neoplasias/metabolismo , Animales , Huesos/metabolismo , Movimiento Celular , Proliferación Celular , Colágeno/química , Exones , Humanos , Glicoproteínas de Membrana/biosíntesis , Ratones , MicroARNs/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Invasividad Neoplásica , Recurrencia Local de Neoplasia , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , ARN Largo no Codificante/metabolismo , Transducción de Señal
16.
Int J Mol Sci ; 21(3)2020 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-31973001

RESUMEN

Escherichia coli is the most widely used protein production host in academia and a major host for industrial protein production. However, recombinant production of eukaryotic proteins in prokaryotes has challenges. One of these is post-translational modifications, including native disulfide bond formation. Proteins containing disulfide bonds have traditionally been made by targeting to the periplasm or by in vitro refolding of proteins made as inclusion bodies. More recently, systems for the production of disulfide-containing proteins in the cytoplasm have been introduced. However, it is unclear if these systems have the capacity for the production of disulfide-rich eukaryotic proteins. To address this question, we tested the capacity of one such system to produce domain constructs, containing up to 44 disulfide bonds, of the mammalian extracellular matrix proteins mucin 2, alpha tectorin, and perlecan. All were successfully produced with purified yields up to 6.5 mg/L. The proteins were further analyzed using a variety of biophysical techniques including circular dichroism spectrometry, thermal stability assay, and mass spectrometry. These analyses indicated that the purified proteins are most likely correctly folded to their native state. This greatly extends the use of E. coli for the production of eukaryotic proteins for structural and functional studies.


Asunto(s)
Citoplasma/metabolismo , Proteínas de Escherichia coli/biosíntesis , Escherichia coli/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Animales , Disulfuros/química , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Proteoglicanos de Heparán Sulfato , Cuerpos de Inclusión/metabolismo , Mucina 2/metabolismo , Periplasma/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica
17.
Int J Cosmet Sci ; 42(4): 388-398, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32453870

RESUMEN

OBJECTIVE: As skin ages, impaired extracellular matrix (ECM) protein synthesis and increased action of degradative enzymes manifest as atrophy, wrinkling and laxity. There is mounting evidence for the functional role of exogenous peptides across many areas, including in offsetting the effects of cutaneous ageing. Here, using an artificial intelligence (AI) approach, we identified peptide RTE62G (pep_RTE62G), a naturally occurring, unmodified peptide with ECM stimulatory properties. The AI-predicted anti-ageing properties of pep_RTE62G were then validated through in vitro, ex vivo and proof of concept clinical testing. METHODS: A deep learning approach was applied to unlock pep_RTE62G from a plant source, Pisum sativum (pea). Cell culture assays of human dermal fibroblasts (HDFs) and keratinocytes (HaCaTs) were subsequently used to evaluate the in vitro effect of pep_RTE62G. Distinct activities such as cell proliferation and ECM protein production properties were determined by ELISA assays. Cell migration was assessed using a wound healing assay, while ECM protein synthesis and gene expression were analysed, respectively, by immunofluorescence microscopy and PCR. Immunohistochemistry of human skin explants was employed to further investigate the induction of ECM proteins by pep_RTE62G ex vivo. Finally, the clinical effect of pep_RTE626 was evaluated in a proof of concept 28-day pilot study. RESULTS: In vitro testing confirmed that pep_RTE62G is an effective multi-functional anti-ageing ingredient. In HaCaTs, pep_RTE62G treatment significantly increases both cellular proliferation and migration. Similarly, in HDFs, pep_RTE62G consistently induced the neosynthesis of ECM protein elastin and collagen, effects that are upheld in human skin explants. Lastly, in our proof of concept clinical study, application of pep_RTE626 over 28 days demonstrated anti-wrinkle and collagen stimulatory potential. CONCLUSION: pep_RTE62G represents a natural, unmodified peptide with AI-predicted and experimentally validated anti-ageing properties. Our results affirm the utility of AI in the discovery of novel, functional topical ingredients.


OBJECTIF: À mesure que la peau vieillit, une altération de la synthèse des protéines de la matrice extracellulaire (ECM) et une action accrue des enzymes dégradantes se manifestent par une atrophie, des rides et un laxisme. Il existe de plus en plus de preuves du rôle fonctionnel des peptides exogènes dans de nombreux domaines, y compris pour compenser les effets du vieillissement cutané. Ici, en utilisant une approche d'intelligence artificielle (AI), nous avons identifié le peptide RTE62G (pep_RTE62G), un peptide naturel non modifié avec des propriétés de stimulation ECM. Les propriétés anti-âge prédites par l'IA de pep_RTE62G ont ensuite été validées par des tests cliniques in vitro, ex vivo et de validation de principe. LES MÉTHODES: Une approche d'apprentissage en profondeur a été appliquée pour déverrouiller pep_RTE62G à partir d'une source végétale, Pisum sativum (pois). Des tests de culture cellulaire de fibroblastes dermiques humains (HDF) et de kératinocytes (HaCaTs) ont ensuite été utilisés pour évaluer l'effet in vitro de pep_RTE62G. Des activités distinctes telles que la prolifération cellulaire et les propriétés de production de protéines ECM ont été déterminées par des tests ELISA. La migration cellulaire a été évaluée à l'aide d'un test de cicatrisation des plaies, tandis que la synthèse des protéines ECM et l'expression des gènes ont été analysées, respectivement, par microscopie à immunofluorescence et PCR. L'immunohistochimie des explants de peau humaine a été utilisée pour approfondir l'induction des protéines ECM par pep_RTE62G ex vivo. Enfin, l'effet clinique de pep_RTE626 a été évalué dans une étude pilote de 28 jours de validation de principe. RÉSULTATS: Les tests in vitro ont confirmé que pep_RTE62G est un ingrédient anti-âge multifonctionnel efficace. Dans HaCaTs, le traitement pep_RTE62G augmente de manière significative à la fois la prolifération et la migration cellulaire. De même, dans les HDF, pep_RTE62G a induit de manière cohérente la néosynthèse de la protéine ECM élastine et collagène, effets qui sont maintenus dans les explants de peau humaine. Enfin, dans notre étude clinique de preuve de concept, l'application de pep_RTE626 sur 28 jours a démontré un potentiel stimulant anti-rides et collagène. CONCLUSION: pep_RTE62G représente un peptide naturel, non modifié avec des propriétés anti-âge prédites par l'IA et validées expérimentalement. Nos résultats confirment l'utilité de l'IA dans la découverte de nouveaux ingrédients topiques fonctionnels.


Asunto(s)
Envejecimiento/efectos de los fármacos , Cosméticos , Aprendizaje Profundo , Queratinocitos/efectos de los fármacos , Péptidos/farmacología , Anciano , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Método Doble Ciego , Proteínas de la Matriz Extracelular/biosíntesis , Femenino , Humanos , Queratinocitos/citología , Persona de Mediana Edad , Pisum sativum/química , Proyectos Piloto , Placebos , Proteínas de Plantas/química , Prueba de Estudio Conceptual , Reproducibilidad de los Resultados
18.
J Mol Cell Cardiol ; 127: 83-96, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30529267

RESUMEN

Cardiac fibrosis (CF), a process characterized by potentiated proliferation of cardiac fibroblasts and excessive secretion and deposition of extracellular matrix (ECM) from the cells, contributes strongly to the pathogenesis of a series of cardiovascular (CV) diseases, including AMI, heart failure and atrial fibrillation. Endothelial-mesenchymal transition (EndMT), one of the sources of transformed cardiac fibroblasts, has been reported as a key factor involved in CF. However, the molecular basis of EndMT has not been thoroughly elucidated to date. At the posttranscriptional level, of the three epigenetic regulators, writer and eraser are reported to be involved in EndMT, but the role of reader in the process is still unknown. In this study, we aimed to explore the role of Bromodomain-containing protein 4 (BRD4), an acetyl-lysine reader protein, in EndMT-induced CF and related mechanisms. We found that BRD4 was upregulated in endothelial cells (ECs) in the pressure-overload mouse heart and that its functional inhibitor JQ1 potently attenuated the TAC-induced CF and preserved cardiac function. In umbilical vein endothelial cells (HUVECs) and mouse aortic endothelial cells (MAECs), bothJQ1 and shRNA-mediated silencing of BRD4 blocked TGF-ß-induced EC migration, EndMT and ECM synthesis and preserved the EC sprouting behavior, possibly through the downregulation of a group of transcription factors specific for EndMT (Snail, Twist and Slug), the Smads pathway and TGF-ß receptor I. In the absence of TGF-ß stimulation, ectopic expression of BRD4 alone could facilitate EndMT, accelerate migration and increase the synthesis of ECM. In vivo, JQ1 also attenuated TAC-induced EndMT and CF, which was consistent with JQ1's intracellular mechanisms of action. Our results showed that BRD4 plays a critical role in EndMT-induced CF and that targeting BRD4 might be a novel therapeutic option for CF.


Asunto(s)
Aorta/patología , Proteínas de Ciclo Celular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Mesodermo/metabolismo , Miocardio/patología , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/efectos adversos , Animales , Biomarcadores/metabolismo , Movimiento Celular/efectos de los fármacos , Constricción , Regulación hacia Abajo/efectos de los fármacos , Proteínas de la Matriz Extracelular/biosíntesis , Fibroblastos/metabolismo , Fibrosis , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo
19.
J Biol Chem ; 293(17): 6603-6610, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29540474

RESUMEN

The visceral (VIS) and subcutaneous (SQ) fat pads are developmentally distinct white adipose tissue depots and contribute differently to inflammation and insulin resistance associated with obesity. The basic helix-loop-helix transcriptional regulator, transcription factor 21 (TCF21), is a marker gene for white adipose tissues and is abundantly expressed in VIS-derived adipose stem cells (ASCs), but not in SQ-derived ASCs. However, TCF21's role in regulating fat depot-specific gene expression and function is incompletely understood. Here, using siRNA-mediated Tcf21 knockdowns and lentiviral gene transfer of TCF21 in mouse ASCs, we demonstrate that TCF21 is required for the VIS ASC-specific expression of interleukin 6 (IL6), a key cytokine that contributes to the proinflammatory nature of VIS depots. Concurrently, TCF21 promotes MMP-dependent collagen degradation and type IV collagen deposition through the regulation of the extracellular matrix (ECM) modifiers, matrix metalloproteinase (MMP) 2, MMP13, and tissue inhibitor of MMP1 (TIMP1), as well as collagen type IV α1 chain (COL4A1) in VIS ASCs. We also found that although IL6 mediates the expression of Mmp13 and Timp1 in VIS ASCs, the TCF21-dependent expression of Mmp2 and Col4a1 is IL6-independent. These results suggest that TCF21 contributes to the proinflammatory environment in VIS fat depots and to active ECM remodeling of these depots by regulating IL6 expression and MMP-dependent ECM remodeling in a spatiotemporally coordinated manner.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Interleucina-6/biosíntesis , Grasa Intraabdominal/metabolismo , Células Madre/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Técnicas de Silenciamiento del Gen , Interleucina-6/genética , Grasa Intraabdominal/citología , Masculino , Ratones , Células Madre/citología
20.
Kidney Int ; 96(5): 1134-1149, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31492508

RESUMEN

Glomerular matrix protein accumulation, mediated largely by mesangial cells, is central to the pathogenesis of diabetic kidney disease. Our previous studies showed that the membrane microdomains caveolae and their marker protein caveolin-1 regulate matrix protein synthesis in mesangial cells in response to diabetogenic stimuli, and that caveolin-1 knockout mice are protected against diabetic kidney disease. In a screen to identify the molecular mechanism underlying this protection, we also established that secreted antifibrotic glycoprotein follistatin is significantly upregulated by caveolin-1 deletion. Follistatin potently neutralizes activins, members of the transforming growth factor-ß superfamily. A role for activins in diabetic kidney disease has not yet been established. Therefore, in vitro, we confirmed the regulation of follistatin by caveolin-1 in primary mesangial cells and showed that follistatin controls both basal and glucose-induced matrix production through activin inhibition. In vivo, we found activin A upregulation by immunohistochemistry in both mouse and human diabetic kidney disease. Importantly, administration of follistatin to type 1 diabetic Akita mice attenuated early diabetic kidney disease, characterized by albuminuria, hyperfiltration, basement membrane thickening, loss of endothelial glycocalyx and podocyte nephrin, and glomerular matrix accumulation. Thus, activin A is an important mediator of high glucose-induced profibrotic responses in mesangial cells, and follistatin may be a potential novel therapy for the prevention of diabetic kidney disease.


Asunto(s)
Activinas/metabolismo , Caveolina 1/metabolismo , Nefropatías Diabéticas/prevención & control , Folistatina/uso terapéutico , Animales , Nefropatías Diabéticas/metabolismo , Evaluación Preclínica de Medicamentos , Proteínas de la Matriz Extracelular/biosíntesis , Folistatina/metabolismo , Masculino , Células Mesangiales/metabolismo , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA