Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 191
Filtrar
1.
Acta Histochem ; 123(5): 151719, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33962151

RESUMEN

Aim of our study was to provide insight into the temporal and spatial expression of FGFR1, FGFR2 and CTGF during normal human lung development which may have an important impact on understanding occurrence of developmental lung anomalies. Morphological parameters were analysed using double immunofluorescence on human embryonal (6th and 7th developmental week-dw) and foetal (8th, 9th and 16th developmental week) human lung samples. FGFR1 and FGFR2 was positive during all the dw in both the epithelium and mesenchyme. The highest number of FGFR1 positive cells was observed during the 6th dw (112/mm2) and 9th dw (87/mm2) in the epithelium compared to the 7th, 8th and 16th dw (Kruskal-Wallis test, p < 0.001, p < 0.0001). The highest number of FGFR1 positive cells in the mesenchyme was observed during the 8th dw (19/mm2) and 16th dw (13/mm2) compared to the 6th, 7th, and 9th dw (Kruskal-Wallis test, p < 0.001, p < 0.0001). The number of FGFR1 positive cells in the epithelium was higher for FGFR2 compared to number of positive cells (Mann-Whitney test, p < 0.0001). FGFR2 showed the highest number in the epithelium during the 7th dw (111/mm2) and 9th dw (87/mm2) compared to 6th, 8th and 16th dw (Kruskal-Wallis test, p < 0.001, p < 0.0001, p < 0.01 respectively). The highest number of FGFR2 positive cells in the mesenchyme was observed during the 9th dw (26/mm2), compared to the 6th, 7th,8th and 16th dw (Kruskal-Wallis test, p < 0.0001), while the number of FGFR2 positive cells in the epithelium was significantly higher than in the mesenchyme (Mann-Whitney test, p < 0.0001). CTGF was negative in both epithelium and mesenchyme during all except the 16th dw in the mesenchyme where it co-localized with FGFR2. FGFR1 and FGFR2 might be essential for epithelial-mesenchymal interactions that determine epithelial branching and mesenchymal growth during early lung development. Sudden increase in FGF1 in the epithelium and FGF2 in the mesenchyme in the foetus at 9th dw could be associated with the onset of foetal breathing movements. CTGF first appear during the foetal lung development.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Pulmón/embriología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Transición Epitelial-Mesenquimal , Epitelio , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Mesodermo , Microscopía Fluorescente
2.
Int J Mol Med ; 48(1)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34036397

RESUMEN

Changes in the dorsal cochlear nucleus (DCN) following exposure to noise play an important role in the development of tinnitus. As the development of several diseases is known to be associated with microRNAs (miRNAs/miRs), the aim of the present study was to identify the miRNAs that may be implicated in pathogenic changes in the DCN, resulting in tinnitus. A previously developed tinnitus animal model was used for this study. The study consisted of four stages, including identification of candidate miRNAs involved in tinnitus development using miRNA microarray analysis, validation of miRNA expression using reverse transcription­quantitative PCR (RT­qPCR), evaluation of the effects of candidate miRNA overexpression on tinnitus development through injection of a candidate miRNA mimic or mimic negative control, and target prediction of candidate miRNAs using mRNA microarray analysis and western blotting. The miRNA microarray and RT­qPCR analyses revealed that miR­375­3p expression was significantly reduced in the tinnitus group compared with that in the non­tinnitus group. Additionally, miR­375­3p overexpression via injection of miR­375­3p mimic reduced the proportion of animals with persistent tinnitus. Based on mRNA microarray and western blot analyses, connective tissue growth factor (CTGF) was identified as a potential target for miR­375­3p. Thus, it was inferred that CTGF downregulation by miR­375­3p may weaken with the decrease in miRNA expression, and the increased pro­apoptotic activity of CTGF may result in more severe neuronal damage, contributing to tinnitus development. These findings are expected to contribute significantly to the development of a novel therapeutic approach to tinnitus, thereby bringing about a significant breakthrough in the treatment of this potentially debilitating condition.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Regulación de la Expresión Génica , MicroARNs/biosíntesis , Acúfeno/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Acúfeno/patología
3.
J Am Heart Assoc ; 10(3): e017633, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33470127

RESUMEN

Background Development of abdominal aortic aneurysm (AAA) is associated with proinflammatory cytokines including interleukin-12 (IL12). Deficiency of interleukin 12p40 (IL12p40) increases localized fibrotic events by promoting TGFß2 (transforming growth factor ß)-dependent anti-inflammatory response. Here, we determined whether IL12p40 deficiency in apolipoprotein E-/- mice attenuates the development of AAA by antagonizing proinflammatory response. Methods and Results Double knockout (DKO) mice were generated by crossbreeding IL12p40-/- mice with apolipoprotein E-/- mice (n=12). Aneurysmal studies were performed using angiotensin II (1 µg/kg/min; subcutaneous). Surprisingly, DKO mice did not prevent the development of AAA with angiotensin II infusion. Immunohistological analysis, however, showed distinct pathological features between apolipoprotein E-/- and DKO mice. Polymerase chain reaction (7 day) and cytokine arrays (28 day) of the aortic tissues from DKO mice showed significantly increased expression of cytokines related to anti-inflammatory response (interleukin 5 and interleukin 13), synthetic vascular smooth muscle cell phenotype (Activin receptor-like kinase-1 (ALK-1), artemin, and betacellulin) and T helper 17-associated response (4-1BB, interleukin-17e (Il17e) and Cd40 ligand (Cd-40L)). Indeed, DKO mice exhibited increased expression of the fibro-proteolytic pathway in the medial layer of aortae induced by cellular communication network factor 2 (CCN2) and Cd3+IL17+ cells compared with apolipoprotein E-/- mice. Laser capture microdissection showed predominant expression of CCN2/TGFß2 in the medial layer of human AAA. Finally, Ccn2 haploinsufficiency in the mice showed decreased AAA incidence in response to elastase infusion, associated with decreased matrix metalloproteinase-2 expression. Conclusions Our study reveals novel roles for IL12p40 deficiency in inducing fibro-proteolytic activities in the aneurysmal mouse model. Mechanistically, these effects of IL12p40 deficiency are mediated by CCN2/matrix metalloproteinase-2 crosstalk in the medial layer of aneurysmal aortae.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/etiología , Factor de Crecimiento del Tejido Conjuntivo/genética , Regulación de la Expresión Génica , Subunidad p40 de la Interleucina-12/deficiencia , Metaloproteinasa 2 de la Matriz/genética , ARN/genética , Anciano , Animales , Aorta Abdominal/diagnóstico por imagen , Aorta Abdominal/fisiopatología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Western Blotting , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Humanos , Subunidad p40 de la Interleucina-12/sangre , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Linfocitos T/metabolismo , Linfocitos T/patología , Ultrasonografía , Rigidez Vascular/fisiología
4.
Biochim Biophys Acta Mol Basis Dis ; 1867(1): 165986, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33065236

RESUMEN

During ischemia/reperfusion (I/R), cardiomyocytes activate pathways that regulate cell survival and death and release factors that modulate fibroblast-to-myofibroblast differentiation. The mechanisms underlying these effects are not fully understood. Polycystin-1 (PC1) is a mechanosensor crucial for cardiac function. This work aims to assess the role of PC1 in cardiomyocyte survival, its role in profibrotic factor expression in cardiomyocytes, and its paracrine effects on I/R-induced cardiac fibroblast function. In vivo and ex vivo I/R and simulated in vitro I/R (sI/R) were induced in wild-type and PC1-knockout (PC1 KO) mice and PC1-knockdown (siPC1) neonatal rat ventricular myocytes (NRVM), respectively. Neonatal rat cardiac fibroblasts (NRCF) were stimulated with conditioned medium (CM) derived from NRVM or siPC1-NRVM supernatant after reperfusion and fibroblast-to-myofibroblast differentiation evaluated. Infarcts were larger in PC1-KO mice subjected to in vivo and ex vivo I/R, and necrosis rates were higher in siPC1-NRVM than control after sI/R. PC1 activated the pro-survival AKT protein during sI/R and induced PC1-AKT-pathway-dependent CTGF expression. Furthermore, conditioned media from sI/R-NRVM induced PC1-dependent fibroblast-to-myofibroblast differentiation in NRCF. This novel evidence shows that PC1 mitigates cardiac damage during I/R, likely through AKT activation, and regulates CTGF expression in cardiomyocytes via AKT. Moreover, PC1-NRVM regulates fibroblast-to-myofibroblast differentiation during sI/R. PC1, therefore, may emerge as a new key regulator of I/R injury-induced cardiac remodeling.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Regulación de la Expresión Génica , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Factor de Crecimiento del Tejido Conjuntivo/genética , Masculino , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPP/genética
5.
Oxid Med Cell Longev ; 2020: 1080168, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33343802

RESUMEN

Heme oxygenase-1 (HO-1) has been shown to exert as an antioxidant and anti-inflammatory enzyme in cardiovascular inflammatory diseases. Flavonoids have been demonstrated to display anti-inflammatory and antioxidant effects through the induction of HO-1. 5,8-Dihydroxy-4',7-dimethoxyflavone (DDF), one of the flavonoid compounds, is isolated from Reevesia formosana. Whether DDF induced HO-1 expression on human cardiac fibroblasts (HCFs) remained unknown. Here, we found that DDF time- and concentration-dependently induced HO-1 protein and mRNA expression, which was attenuated by pretreatment with reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC) in HCFs. DDF-enhanced ROS generation was attenuated by NAC, but not by either diphenyleneiodonium chloride (DPI, Nox inhibitor) or MitoTempol (mitochondrial ROS scavenger). Interestingly, pretreatment with glutathione (GSH) inhibited DDF-induced HO-1 expression. The ratio of GSH/GSSG was time-dependently decreased in DDF-treated HCFs. DDF-induced HO-1 expression was attenuated by an inhibitor of p38 MAPK (p38i VIII) or siRNA, but not by MEK1/2 (PD98059) or JNK1/2 (SP600125). DDF-stimulated p38 MAPK phosphorylation was inhibited by GSH or p38i VIII. Moreover, DDF-induced HO-1 expression was mediated through Nrf2 phosphorylation and translocation into the nucleus which was attenuated by NAC or p38 siRNA. DDF also stimulated antioxidant response element (ARE) promoter activity which was inhibited by NAC, GSH, or p38i VIII. Interaction between Nrf2 and the ARE-binding sites on the HO-1 promoter was revealed by chromatin immunoprecipitation assay, which was attenuated by NAC, GSH, or p38i VIII. We further evaluated the functional effect of HO-1 expression on the thrombin-induced fibrotic responses. Our result indicated that the induction of HO-1 by DDF can attenuate the thrombin-induced connective tissue growth factor expression. These results suggested that DDF-induced HO-1 expression is, at least, mediated through the activation of the ROS-dependent p38 MAPK/Nrf2 signaling pathway in HCFs. Thus, the upregulation of HO-1 by DDF could be a candidate for the treatment of heart fibrosis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Fibroblastos/metabolismo , Flavonas/farmacología , Hemo-Oxigenasa 1/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocardio/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trombina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Inducción Enzimática/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Humanos , Factor 2 Relacionado con NF-E2/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
6.
Appl Microbiol Biotechnol ; 104(23): 9979-9990, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33074415

RESUMEN

Human connective tissue growth factor (CTGF) is a secreted cysteine-rich peptide that stimulates cell proliferation, migration, and extracellular matrix production during tissue development, differentiation, angiogenesis, implantation, wound healing, and fibrosis processes, with broad application in the medical and cosmetic medical fields. However, the production of CTGF is currently limited by its low yield and purity in current bioreactors. In this study, two genetically modified silkworm strains were generated harboring artificially designed CTGF-8ht and pepCTGF-8ht genes, respectively, that contain an enhanced His-tag with eight histidine residues with or without a transdermal peptide (pep). Both recombinant CTGF-8ht and pepCTGF-8ht proteins were successfully expressed in the silkworm silk gland and cocoon, and could be easily extracted and purified from the cocoon by single-affinity immunoprecipitation column chromatography, achieving a purity of more than 95%. Moreover, compared with CTGF-8ht protein, pepCTGF-8ht protein exhibited better cell proliferation activity by activating the extracellular signal-regulated kinase (ERK) pathway and enhanced hyaluronic acid synthesis activity by upregulating hyaluronan synthase 3 expression; moreover, the addition of pep significantly improved the transmembrane ability of CTGF-8ht protein. These results should help to promote the application prospects of CTGF and further guide the design and development of protein drugs from silkworm and other bioreactor systems. KEY POINTS : A silkworm bioreactor was optimized to produce connective tissue growth factor (CTGF) The transgene contained an enhanced 8-His-tag and transmembrane peptide (pep) Recombinant CTGF was easily purified with maintained or higher biological activity.


Asunto(s)
Reactores Biológicos , Bombyx , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Animales , Proliferación Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Humanos , Ácido Hialurónico , Proteínas Recombinantes de Fusión/biosíntesis , Seda
7.
Oncogene ; 39(22): 4375-4389, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32313226

RESUMEN

Hippo signaling functions to limit cellular growth, but the aberrant nuclear accumulation of its downstream YAP1 leads to carcinogenesis. YAP1/TEAD complex activates the oncogenic downstream transcription, such as CTGF and c-Myc. How YAP1 is protected in the cytoplasm from ubiquitin-mediated degradation remains elusive. In this study, a member of Angiomotin (Motin) family, AMOTL1 (Angiomotin Like 1), was screened out as the only one to promote YAP1 nuclear accumulation by several clinical cohorts, which was further confirmed by the cellular functional assays. The interaction between YAP1 and AMOTL1 was suggested by co-immunoprecipitation and immunofluorescent staining. The clinical significance of the AMOTL1-YAP1-CTGF axis in gastric cancer (GC) was analyzed by multiple clinical cohorts. Moreover, the therapeutic effect of targeting the oncogenic axis was appraised by drug-sensitivity tests and xenograft-formation assays. The upregulation of AMOTL1 is associated with unfavorable clinical outcomes of GC, and knocking down AMOTL1 impairs its oncogenic properties. The cytoplasmic interaction between AMOTL1 and YAP1 protects each other from ubiquitin-mediated degradation. AMOTL1 promotes YAP1 translocation into the nuclei to activate the downstream expression, such as CTGF. Knocking down AMOTL1, YAP1, and CTGF enhances the therapeutic efficacies of the first-line anticancer drugs. Taken together, AMOTL1 plays an oncogenic role in gastric carcinogenesis through interacting with YAP1 and promoting its nuclear accumulation. A combination of AMOTL1, YAP1, and CTGF expression might serve as a surrogate of Hippo activation status. The co-activation of the AMOTL1/YAP1-CTGF axis is associated with poor clinical outcomes of GC patients, and targeting this oncogenic axis may enhance the chemotherapeutic effects.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Transformación Celular Neoplásica/genética , Proteínas de la Membrana/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias Gástricas/genética , Factores de Transcripción/fisiología , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales/genética , Angiomotinas , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Factor de Crecimiento del Tejido Conjuntivo/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Xenoinjertos , Vía de Señalización Hippo , Humanos , Estimación de Kaplan-Meier , Proteínas de la Membrana/genética , Ratones , Proteínas de Neoplasias/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes/metabolismo , Transducción de Señal , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Factores de Transcripción/genética , Verteporfina/farmacología , Verteporfina/uso terapéutico , Proteínas Señalizadoras YAP
8.
Nanoscale ; 12(11): 6385-6393, 2020 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-32134425

RESUMEN

Skin fibrosis occurs in a variety of human diseases but the current anti-fibrosis treatments are not sufficient. One major cause of fibrotic diseases shared across diverse organ fibrosis is uncontrolled overexpression of the connective tissue growth factor (CTGF, also known as CCN2). Here, we examine the anti-fibrotic activity of RNAi therapy utilizing siRNA against CTGF with a new drug delivery system (DDS), 'DegradaBALL', which is based on porous nanoparticles, for durable CTGF gene silencing. DegradaBALL is a modular DDS having many favorable properties for RNA delivery such as effective intracellular uptake, convenient drug loading, biocompatibility, sustained release profile and biodegradability. DegradaBALL loaded with siCTGF, named 'LEM-S401', showed highly durable and effective CTGF gene-silencing in TGF-ß induced lung fibrosis and skin fibrosis model cells, A549 and HaCaT, respectively. In addition, LEM-S401 induced knockdown of collagen types I and III, which are excess extracellular matrix components in fibrotic skin in addition to CTGF in the mouse wound healing model. Most importantly, we showed that LEM-S401 effectively inhibited the formation of hypertrophic scars in wound-associated dermal fibrosis mouse models, during both the epidermis recovery and tissue remodeling process. Our findings suggest that LEM-S401 could be a highly potent therapeutic option for skin fibrotic diseases.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo , Interferencia de ARN , ARN Interferente Pequeño , Enfermedades de la Piel , Piel/metabolismo , Células A549 , Animales , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Colágeno Tipo III/biosíntesis , Colágeno Tipo III/genética , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Factor de Crecimiento del Tejido Conjuntivo/genética , Fibrosis , Humanos , Masculino , Ratones , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Piel/patología , Enfermedades de la Piel/genética , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología , Enfermedades de la Piel/terapia
9.
Cell Tissue Res ; 379(3): 549-559, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31773302

RESUMEN

Diabetic nephropathy (DN) is one of the most devastating complications of diabetes. Connective tissue growth factor (CTGF) levels are up-regulated in patients with DN and in renal tubular epithelial cells (RTECs) exposed to high glucose (HG). The underlying epigenetic mechanism remains to be elucidated. In the present study, we investigate the role of myocardin-related transcription factor A (MRTF-A) in HG-induced CTGF transcription in RTECs. We report that in two different animal models of DN, one induced by streptozotocin (STZ) injection and the other induced by high-fat diet (HFD) feeding, MRTF-A deficiency attenuated CTGF induction in the kidneys. In cultured RTECs, MRTF-A knockdown similarly ameliorated CTGF induction by HG treatment. Upon CTGF induction, there was an increase in acetylated histone H3 (AcH3) and trimethylated H3K4 (H3K4Me3) on the CTGF promoter region accompanying a decrease in dimethylated H3K9 (H3K9Me2). MRTF-A ablation in vivo or depletion in vitro comparably dampened the accumulation of AcH3 and H3K4Me3 but restored H3K9Me2 on the CTGF promoter. Further analyses revealed that MRTF-A interacted with and recruited histone demethylase KDM3A to the CTGF promoter to activate transcription. KDM3A silencing equivalently weakened HG-induced CTGF induction in RTECs. In conclusion, MRTF-A contributes to HG-induced CTGF transcription via an epigenetic mechanism.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Nefropatías Diabéticas/metabolismo , Transactivadores/metabolismo , Animales , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Nefropatías Diabéticas/genética , Modelos Animales de Enfermedad , Epigenómica/métodos , Células Epiteliales/metabolismo , Glucosa/administración & dosificación , Glucosa/metabolismo , Células HEK293 , Humanos , Túbulos Renales/citología , Túbulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transactivadores/deficiencia , Transactivadores/genética , Transcripción Genética , Regulación hacia Arriba
10.
Mol Med Rep ; 21(1): 181-190, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31746398

RESUMEN

Connective tissue growth factor (CTGF) is a possible key determinant of progressive fibrosis. Nanotechnology has been considered as a potential tool for developing novel drug delivery systems for various diseases, including liver fibrosis. The present study aimed to investigate the potential antifibrotic activity of CTGF small interfering RNA (siRNA) mediated by polyethyleneimine (PEI)­functionalized magnetic iron oxide (Fe3O4) nanoparticles (NPs) in LX­2 cells. PEI­Fe3O4/siRNA complexes were synthesized to facilitate siRNA delivery and were transfected into LX­2 cells. Laser confocal microscopy was employed to investigate the cell uptake of PEI­Fe3O4/siRNA complexes. Reverse transcription­quantitative PCR (RT­qPCR) and western blotting were used to verify the effect of gene silencing. The results showed that siRNA­loaded PEI­Fe3O4 exhibited low cytotoxicity. The transfection efficiency of PEI­Fe3O4/siRNA reached 73.8%, and RT­qPCR and western blotting demonstrated effective gene silencing. These results indicated that CTGF siRNA delivered by PEI­Fe3O4 NPs significantly reduces CTGF expression and collagen production in activated LX­2 cells, providing a basis for future in vivo studies.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Silenciador del Gen , Marcación de Gen , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/metabolismo , Nanopartículas de Magnetita/química , ARN Interferente Pequeño/farmacología , Transfección , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/terapia , ARN Interferente Pequeño/genética
11.
Biochem Biophys Res Commun ; 519(2): 309-315, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31506177

RESUMEN

Jiadifenolide has been reported to have neurotrophin-like activity in primary rat cortical neurons, and also possesses neurotrophic effects in neuronal precursor cells derived from human induced pluripotent stem cells (hiPSCs), as we have previously reported. However, the molecular mechanisms by which jiadifenolide exerts its neurotrophic effects in rat and human neurons are unknown. Thus, we aimed to investigate the molecular mechanisms and pathways by which jiadifenolide promotes neurotrophic effects. Here, we found that jiadifenolide activated cellular communication network factor (CCN) signaling pathways by up-regulating mRNA level expression of CCN genes in human neuronal cells. We also found that CCN2 (also known as connective tissue growth factor, CTGF) protein promotes neurotrophic effects through activation of the p44/42 mitogen-activated protein kinase signaling pathway. This is the first discovery which links neurotrophic activity with CCN signaling.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Sesquiterpenos/farmacología , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Sesquiterpenos/síntesis química , Sesquiterpenos/química
12.
Circ Res ; 125(9): 834-846, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31495264

RESUMEN

RATIONALE: Pathogenic variations in the lamin gene (LMNA) cause familial dilated cardiomyopathy (DCM). LMNA insufficiency caused by LMNA pathogenic variants is believed to be the basic mechanism underpinning LMNA-related DCM. OBJECTIVE: To assess whether silencing of cardiac Lmna causes DCM and investigate the role of Yin Yang 1 (Yy1) in suppressing Lmna DCM. METHODS AND RESULTS: We developed a Lmna DCM mouse model induced by cardiac-specific Lmna short hairpin RNA. Silencing of cardiac Lmna induced DCM with associated cardiac fibrosis and inflammation. We demonstrated that upregulation of Yy1 suppressed Lmna DCM and cardiac fibrosis by inducing Bmp7 expression and preventing upregulation of Ctgf. Knockdown of upregulated Bmp7 attenuated the suppressive effect of Yy1 on DCM and cardiac fibrosis. However, upregulation of Bmp7 alone was not sufficient to suppress DCM and cardiac fibrosis. Importantly, upregulation of Bmp7 together with Ctgf silencing significantly suppressed DCM and cardiac fibrosis. Mechanistically, upregulation of Yy1 regulated Bmp7 and Ctgf reporter activities and modulated Bmp7 and Ctgf gene expression in cardiomyocytes. Downregulation of Ctgf inhibited TGF-ß (transforming growth factor-ß)/Smad signaling in DCM hearts. Regulation of both Bmp7 and Ctgf further suppressed TGFß/Smad signaling. In addition, co-modulation of Bmp7 and Ctgf reduced CD3+ T cell numbers in DCM hearts. CONCLUSIONS: Our findings demonstrate that upregulation of Yy1 or co-modulation of Bmp7 and Ctgf offer novel therapeutic strategies for the treatment of DCM caused by LMNA insufficiency.


Asunto(s)
Proteína Morfogenética Ósea 7/biosíntesis , Cardiomiopatías/metabolismo , Cardiomiopatías/prevención & control , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Factor de Transcripción YY1/biosíntesis , Animales , Proteína Morfogenética Ósea 7/genética , Cardiomiopatías/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Endotelio Vascular/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción YY1/genética
13.
FASEB J ; 33(11): 12554-12564, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31451010

RESUMEN

Mycobacterium tuberculosis (M.tb) infection in lung causes pulmonary fibrosis, which leads to the irreversible reduction of pulmonary function. Fibrotic protein connective tissue growth factor (CTGF) expression has been confirmed to play a crucial role in lung fibrosis. However, the underlying signal pathway and effect of M.tb on CTGF expression in human lung fibroblasts are unclear. Our results revaled that M.tb caused time- and concentration-dependent increases in CTGF expression in human lung fibroblasts. A mechanistic investigation revealed that M.tb induced CTGF expression through TLR2 but not TLR4. The promoter activity assay indicated that M.tb-induced CTGF activity was mainly controlled by the promoter region at -747 to -184 bp, which contained signal transducer and activator of transcription 3 and activator protein 1 (AP-1) binding sites. Moreover, curcumin (AP-1 inhibitor) restrained M.tb-induced CTGF expression. M.tb also induced increases in AP-1 luciferase activity and DNA binding activity of c-Jun and c-Fos on the CTGF promoter. Furthermore, the knockdown of c-Jun by small interfering RNA attenuated M.tb-induced CTGF expression and AP-1 luciferase activity. A JNK inhibitor (SP600125) and a JNK dominant-negative mutant suppressed M.tb-induced CTGF expression. We also discovered that M.tb could induce the phosphorylation of JNK and c-Jun. Furthermore, SP600125 inhibited M.tb-induced c-Jun phosphorylation and AP-1- luciferase activity. M.tb-induced fibronectin expression was inhibited by anti-CTGF antibody. These results demonstrate that M.tb is activated through TLR2 to induce JNK activation, further increasing the DNA binding activity of c-Jun and c-Fos and finally inducing CTGF expression and extracellular matrix production.-Lee, H.-S., Hua, H.-S., Wang, C.-H., Yu, M.-C., Chen, B.-C., Lin, C.-H. Mycobacterium tuberculosis induces connective tissue growth factor expression through the TLR2-JNK-AP-1 pathway in human lung fibroblasts.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Fibroblastos/metabolismo , Pulmón/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Mycobacterium tuberculosis/metabolismo , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Factor de Transcripción AP-1/metabolismo , Tuberculosis Pulmonar/metabolismo , Antracenos/farmacología , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/patología , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Elementos de Respuesta , Tuberculosis Pulmonar/patología
14.
Am J Physiol Renal Physiol ; 317(4): F839-F851, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31364374

RESUMEN

The small GTPase Rho and its effector Rho kinase (ROCK) are involved in the pathogenesis of diabetic kidney disease. Rho kinase has two isoforms: ROCK1 and ROCK2. However, it remains unclear which is mainly involved in the progression of diabetic glomerulosclerosis and the regulation of profibrotic mediators. Glomeruli isolated from type 2 diabetic db/db mice demonstrated increased gene expression of transforming growth factor (TGF)-ß and its downstream profibrotic mediators. Chemical inhibition of ROCK suppressed the expression of profibrotic mediators in both isolated glomeruli and cultured mesangial cells. An investigation of mechanisms underlying this observation revealed activated ROCK functions through the phosphorylation of JNK and Erk and the nuclear translocation of NF-κB via actin dynamics. Knockdown by siRNA against ROCK1 and ROCK2 showed that ROCK2 but not ROCK1 controls this fibrotic machinery. Further in vivo experiments showed that ROCK2 activity in the renal cortex of db/db mice was elevated compared with control db/m mice. Importantly, oral administration of ROCK2 inhibitor attenuated renal ROCK2 activity, albuminuria, and glomerular fibrosis in db/db mice. These observations indicate that ROCK2 is a key player in the development of diabetic renal injury. Glomerular ROCK2 may be a potential therapeutic target for the treatment of diabetic kidney disease.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Citoesqueleto/metabolismo , Fibrosis/genética , Mesangio Glomerular/metabolismo , FN-kappa B/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Quinasas Asociadas a rho/metabolismo , Actinas/metabolismo , Animales , Nefropatías Diabéticas/metabolismo , Activación Enzimática , Mesangio Glomerular/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos NOD , Quinasas Asociadas a rho/antagonistas & inhibidores
15.
PLoS One ; 14(7): e0218068, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31314805

RESUMEN

Oro-facial fibrosis in systemic sclerosis (Scleroderma;SSc) has a major impact on mouth function, facial appearance, and patient quality of life. Lipotransfer is a method of reconstruction that can be used in the treatment of oro-facial fibrosis. The effect of this treatment not only restores oro-facial volume but has also been found to reverse the effects of oro-facial fibrosis. Adipose derived stem cells (ADSCs) within the engrafted adipose tissue have been shown to be anti-fibrotic in SSc and are proposed as the mechanism of the anti-fibrotic effect of lipotransfer. A cohort of 62 SSc patients with oro-facial fibrosis were assessed before and after stem cell enriched lipotransfer treatment. Clinical evaluation included assessment of mouth function using a validated assessment tool (Mouth Handicap in Systemic Sclerosis Scale-MHISS), validated psychological measurements and pre and post-operative volumetric assessment. In addition, to understand the mechanism by which the anti-fibrotic effect of ADSCs occur, SSc derived fibroblasts and ADSCs from this cohort of patients were co-cultured in direct and indirect culture systems and compared to monoculture controls. Cell viability, DNA content, protein secretion of known fibrotic mediators including growth factor- ß1 (TGF ß-1) and connective tissue growth factor (CTGF) using ELISA analysis and fibrosis gene expression using a fibrosis pathway specific qPCR array were evaluated. Mouth function (MHISS) was significantly improved (6.85±5.07) (p<0.0001) after treatment. All psychological measures were significantly improved: DAS 24 (12.1±9.5) (p<0.0001); HADS-anxiety (2.8±3.2) (p<0.0001), HADS-depression (2.0±3.1) (p<0.0001); BFNE (2.9 ± 4.3) (p<0.0001); VAS (3.56±4.1) (p<0.0001). Multiple treatments further improved mouth function (p<0.05), DAS (p<0.0001) and VAS (p = 0.01) scores. SSc fibroblast viability and proliferation was significantly reduced in co-culture compared to monoculture via a paracrine effect over 14 days (p < 0.0001). Protein secretion of transforming growth factor (TGF-ß1) and connective tissue growth factor (CTGF) was significantly reduced in co-culture compared to monoculture (p < 0.0001). Multiple fibrosis associated genes were down regulated in SSc co-culture compared to monoculture after 14 days including Matrix metalloproteinase-8 (MMMP-8), Platelet derived growth factor-ß (PDGF-ß) and Integrin Subunit Beta 6 (ITG-ß6). Autologous stem cell enriched lipotransfer significantly improved the effects of oro-facial fibrosis in SSc in this open cohort study. Lipotransfer may reduce dermal fibrosis through the suppression of fibroblast proliferation and key regulators of fibrogenesis including TG-ß1 and CTGF. Our findings warrant further investigation in a randomised controlled trial.


Asunto(s)
Tejido Adiposo , Fibroblastos , Recuperación de la Función , Esclerodermia Sistémica , Células Madre , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Tejido Adiposo/trasplante , Anciano , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Femenino , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibrosis , Regulación de la Expresión Génica , Humanos , Cadenas beta de Integrinas/biosíntesis , Masculino , Metaloproteinasa 8 de la Matriz/biosíntesis , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-sis/biosíntesis , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Esclerodermia Sistémica/terapia , Células Madre/metabolismo , Células Madre/patología , Factor de Crecimiento Transformador beta1/biosíntesis
16.
PLoS One ; 14(6): e0218178, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31170244

RESUMEN

The role of the microenvironment in driving connective tissue disease is being increasingly appreciated. Matricellular proteins of the CCN family are signaling modifiers that are secreted by cells into the extracellular matrix microenvironment where they have profound, context-dependent effects on organ development, homeostasis and disease. Indeed, CCN proteins are emergent targets for therapeutic intervention. Recent evidence suggests that, in vivo, CCN3 has effects opposing CCN2. Moreover, when CCN3 expression is high, CCN2 expression is low. That is, they appear to be regulated in a yin/yang fashion, leading to the hypothesis that the CCN2:CCN3 ratio is important to control tissue homeostasis. To begin to test the hypothesis that alterations in CCN2:CCN3 expression might be important in skin biology in vivo, we evaluated the relative ex vivo effects of the profibrotic protein TGFbeta1 on dermal fibroblasts on protein and RNA expression of CCN3 and CCN2, as well as the related protein CCN1. We also used signal transduction inhibitors to begin to identify the signal transduction pathways controlling the ability of fibroblasts to respond to TGFbeta1. As anticipated, CCN1 and CCN2 protein and mRNA were induced by TGFbeta1 in human dermal fibroblasts. This induction was blocked by TAK1, FAK, YAP1 and MEK inhibition. Conversely, TGFbeta1 suppressed CCN3 mRNA expression in a fashion insensitive to FAK, MEK, TAK1 or YAP1 inhibition. Unexpectedly, CCN3 protein was not detected in human dermal fibroblasts basally. These data suggest that, in dermal fibroblasts, the profibrotic protein TGFbeta1 has a divergent effect on CCN3 relative to CCN2 and CCN1, both at the mRNA and protein level. Given that the major source in skin in vivo of CCN proteins are fibroblasts, our data are consistent that alterations in CCN2/CCN1: CCN3 ratios in response to profibrotic agents such as TGFbeta1 may play a role in connective tissue pathologies including fibrosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Proteína 61 Rica en Cisteína/biosíntesis , Quinasa 1 de Adhesión Focal/metabolismo , Regulación de la Expresión Génica , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína Hiperexpresada del Nefroblastoma/biosíntesis , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Dermis , Fibroblastos , Humanos , Proteínas Señalizadoras YAP
17.
Obesity (Silver Spring) ; 27(6): 957-962, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31004409

RESUMEN

OBJECTIVE: Connective tissue growth factor (CTGF) is an important regulator of fibrogenesis in many organs. This study evaluated the interrelationship among adipose tissue CTGF expression, fat mass, and insulin resistance in humans. METHODS: This study examined (1) CTGF gene expression in human subcutaneous preadipocytes before and after inducing adipogenesis; (2) relationships among abdominal subcutaneous adipose tissue CTGF gene expression, body fat mass, and indices of insulin sensitivity, including the hepatic insulin sensitivity index and the hyperinsulinemic-euglycemic clamp procedure in conjunction with stable isotope glucose tracer infusion, in 72 people who had marked differences in adiposity and insulin sensitivity; (3) localization of CTGF protein in subcutaneous adipose tissue; and (4) effect of progressive (5%, 11%, and 16%) weight loss on adipose tissue CTGF gene expression. RESULTS: CTGF was highly expressed in preadipocytes, not adipocytes. Adipose tissue CTGF expression was strongly correlated with body fat mass and both skeletal muscle and liver insulin sensitivity, and CTGF-positive cells were predominantly found in areas of fibrosis. Progressive weight loss caused a stepwise decrease in adipose tissue CTGF expression. CONCLUSIONS: It was concluded that increased CTGF expression is associated with adipose tissue expansion, adipose tissue fibrosis, and multi-organ insulin resistance in people with obesity.


Asunto(s)
Adiposidad/fisiología , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Adulto , Anciano , Factor de Crecimiento del Tejido Conjuntivo/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Obesidad/genética , Adulto Joven
18.
Eur J Heart Fail ; 21(4): 482-491, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30675967

RESUMEN

AIMS: Atrial fibrillation is the most prevalent sustained arrhythmia associated with arrhythmic ventricular contractions, incident heart failure, increased morbidity and mortality. The relationship between arrhythmic contractions and ventricular remodelling is incompletely understood. The aim of this study was to characterize the influence of irregular contractions on pro-fibrotic signalling in neonatal rat ventricular cardiomyocytes (NRVM). METHODS AND RESULTS: Neonatal rat ventricular cardiomyocytes were paced via field stimulation at 3 Hz for 24 h. Irregularity was created by pseudorandomized variation of stimulation intervals and compared to regular pacing. Treatment of neonatal cardiac fibroblasts (NCF) with medium of irregularly paced NRVM increased protein expression of collagen I (206 ± 62%, P = 0.0121) and collagen III (51 ± 37%, P = 0.0119). To identify the underlying mechanism, expression of pro-fibrotic connective tissue growth factor (CTGF) and transforming growth factor beta (TGF-ß) was assessed. In irregularly paced NRVM, increased protein expression of CTGF (80 ± 22%, P = 0.0035) and TGF-ß (122 ± 31%, P = 0.0022) was associated with enhanced excretion of both proteins into the medium. Electron paramagnetic resonance spectroscopy revealed an increased production of reactive oxygen species (46 ± 21%, P = 0.0352) after irregular pacing accompanied by increased 8-hydroxydeoxyguanosine staining (214 ± 53%, P = 0.0011). Irregular pacing was associated with elevated mRNA levels of anti-oxidative superoxide dismutase 1 (25 ± 7%, P = 0.0175), superoxide dismutase 3 (20 ± 7%, P = 0.0309), and catalase (20 ± 7%, P = 0.046). CONCLUSION: These data demonstrate that irregular pacing is an important inductor of pro-fibrotic signalling in NRVM involving paracrine effects of CTGF and TGF-ß as well as increased oxidative stress. Thus, irregularity of the heart beat might directly be involved in the progression of maladaptive remodelling processes in atrial fibrillation.


Asunto(s)
Fibrilación Atrial/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Fibrosis/metabolismo , Miocitos Cardíacos/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Fibrilación Atrial/fisiopatología , Células Cultivadas , Femenino , Colágenos Fibrilares/biosíntesis , Fibroblastos/metabolismo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Masculino , Estrés Oxidativo , Comunicación Paracrina , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Transducción de Señal
19.
FASEB J ; 33(2): 2047-2057, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30216109

RESUMEN

In skeletal muscle, extracellular matrix (ECM) remodeling can either support the complete regeneration of injured muscle or facilitate pathologic fibrosis and muscle degeneration. Muscular dystrophy (MD) is a group of genetic disorders that results in a progressive decline in muscle function and is characterized by the abundant deposition of fibrotic tissue. Unlike acute injury, where ECM remodeling is acute and transient, in MD, remodeling persists until fibrosis obstructs the regenerative efforts of diseased muscles. Thus, understanding how ECM is deposited and organized is critical in the context of muscle repair. Connective tissue growth factor (CTGF or CCN2) is a matricellular protein expressed by multiple cell types in response to tissue injury. Although used as a general marker of fibrosis, the cell type-dependent role of CTGF in dystrophic muscle has not been elucidated. To address this question, a conditional Ctgf myofiber and fibroblast-knockout mouse lines were generated and crossed to a dystrophic background. Only myofiber-selective inhibition of CTGF protected δ-sarcoglycan-null ( Sgcd-/-) mice from the dystrophic phenotype, and it did so by affecting collagen organization in a way that allowed for improvements in dystrophic muscle regeneration and function. To confirm that muscle-specific CTGF functions to mediate collagen organization, we generated mice with transgenic muscle-specific overexpression of CTGF. Again, genetic modulation of CTGF in muscle was not sufficient to drive fibrosis, but altered collagen content and organization after injury. Our results show that the myofibers are critical mediators of the deleterious effects associated with CTGF in MD and acutely injured skeletal muscle.-Petrosino, J. M., Leask, A., Accornero, F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo , Matriz Extracelular , Regulación de la Expresión Génica , Fibras Musculares Esqueléticas , Distrofia Muscular Animal , Animales , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Factor de Crecimiento del Tejido Conjuntivo/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosis , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Sarcoglicanos/deficiencia
20.
Retina ; 39(2): 265-273, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29190236

RESUMEN

PURPOSE: We sought to characterize the angiofibrotic and apoptotic effects of vascular endothelial growth factor (VEGF)-inhibition on fibrovascular epiretinal membranes in eyes with traction retinal detachment because of proliferative diabetic retinopathy. METHODS: Membranes were excised from 20 eyes of 19 patients (10 randomized to intravitreal bevacizumab, 10 controls) at vitrectomy. Membranes were stained with antibodies targeting connective tissue growth factor (CTGF) or VEGF and colabeled with antibodies directed against endothelial cells (CD31), myofibroblasts, or retinal pigment epithelium markers. Quantitative and colocalization analyses of antibody labeling were obtained through immunofluorescence confocal microscopy. Masson trichrome staining, cell counting of hematoxylin and eosin sections, and terminal dUTP nick-end labeling staining were performed. RESULTS: High levels of fibrosis were observed in both groups. Cell apoptosis was higher (P = 0.05) in bevacizumab-treated membranes compared with controls. The bevacizumab group had a nonsignificant reduction in colocalization in CD31-CTGF and cytokeratin-VEGF studies compared with controls. Vascular endothelial growth factor in extracted membranes was positively correlated with vitreous levels of VEGF; CTGF in extracted membranes was negatively correlated with vitreous levels of CTGF. CONCLUSION: Bevacizumab suppresses vitreous VEGF levels, but does not significantly alter VEGF or CTGF in diabetic membranes that may be explained by high baseline levels of fibrosis. Bevacizumab may cause apoptosis within fibrovascular membranes.


Asunto(s)
Apoptosis , Bevacizumab/administración & dosificación , Retinopatía Diabética/patología , Membrana Epirretinal/cirugía , Retina/patología , Vitrectomía/métodos , Actinas/biosíntesis , Inhibidores de la Angiogénesis/administración & dosificación , Proliferación Celular , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Retinopatía Diabética/complicaciones , Retinopatía Diabética/tratamiento farmacológico , Membrana Epirretinal/complicaciones , Membrana Epirretinal/patología , Fibrosis/patología , Humanos , Inyecciones Intravítreas , Queratinas/biosíntesis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Estudios Prospectivos , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...