Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(3): e0299820, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38507437

RESUMO

Targeting endolysosomes is a strategy extensively pursued for treating cancers, including glioblastomas (GBMs), on the basis that the intact function of these subcellular organelles is key to tumor cell autophagy and survival. Through gene expression analyses and cell type abundance estimation in GBMs, we showed that genes associated with the endolysosomal machinery are more prominently featured in non-tumor cells in GBMs than in tumor cells, and that tumor-associated macrophages represent the primary immune cell type that contributes to this trend. Further analyses found an enrichment of endolysosomal pathway genes in immunosuppressive (pro-tumorigenic) macrophages, such as M2-like macrophages or those associated with worse prognosis in glioma patients, but not in those linked to inflammation (anti-tumorigenic). Specifically, genes critical to the hydrolysis function of endolysosomes, including progranulin and cathepsins, were among the most positively correlated with immunosuppressive macrophages, and elevated expression of these genes is associated with worse patient survival in GBMs. Together, these results implicate the hydrolysis function of endolysosomes in shaping the immunosuppressive microenvironment of GBM. We propose that targeting endolysosomes, in addition to its detrimental effects on tumor cells, can be leveraged for modulating immunosuppression to render GBMs more amenable to immunotherapies.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioblastoma/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Microambiente Tumoral/genética
2.
Neuro Oncol ; 25(9): 1563-1575, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-36689342

RESUMO

BACKGROUND: Telomere maintenance mechanisms are required to enable the replicative immortality of malignant cells. While most cancers activate the enzyme telomerase, a subset of cancers uses telomerase-independent mechanisms termed alternative lengthening of telomeres (ALT). ALT occurs via homology-directed-repair mechanisms and is frequently associated with ATRX mutations. We previously showed that a subset of adult glioblastoma (GBM) patients with ATRX-expressing ALT-positive tumors harbored loss-of-function mutations in the SMARCAL1 gene, which encodes an annealing helicase involved in replication fork remodeling and the resolution of replication stress. However, the causative relationship between SMARCAL1 deficiency, tumorigenesis, and de novo telomere synthesis is not understood. METHODS: We used a patient-derived ALT-positive GBM cell line with native SMARCAL1 deficiency to investigate the role of SMARCAL1 in ALT-mediated de novo telomere synthesis, replication stress, and gliomagenesis in vivo. RESULTS: Inducible rescue of SMARCAL1 expression suppresses ALT indicators and inhibits de novo telomere synthesis in GBM and osteosarcoma cells, suggesting that SMARCAL1 deficiency plays a functional role in ALT induction in cancers that natively lack SMARCAL1 function. SMARCAL1-deficient ALT-positive cells can be serially propagated in vivo in the absence of detectable telomerase activity, demonstrating that the SMARCAL1-deficient ALT phenotype maintains telomeres in a manner that promotes tumorigenesis. CONCLUSIONS: SMARCAL1 deficiency is permissive to ALT and promotes gliomagenesis. Inducible rescue of SMARCAL1 in ALT-positive cell lines permits the dynamic modulation of ALT activity, which will be valuable for future studies aimed at understanding the mechanisms of ALT and identifying novel anticancer therapeutics that target the ALT phenotype.


Assuntos
Glioblastoma , Telomerase , Humanos , Telomerase/genética , Telomerase/metabolismo , Glioblastoma/genética , Homeostase do Telômero , Mutação , Telômero/genética , Telômero/metabolismo , Carcinogênese , Transformação Celular Neoplásica/genética , DNA Helicases/genética , DNA Helicases/metabolismo
3.
J Ovarian Res ; 15(1): 114, 2022 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-36266675

RESUMO

BACKGROUND: Epithelial ovarian cancer (OC) is the most lethal gynecological malignancy and patients present with significant metastatic burden, particularly to the adipose-rich microenvironment of the omentum. Recent evidence has highlighted the importance of metabolic adaptations in enabling this metastasis, leading to significant interest in evolving the arsenal of tools used to study OC metabolism. In this study, we demonstrate the capability of genetically encoded fluorescent biosensors to study OC, with a focus on 3D organoid models that better recapitulate in vivo tumor microenvironments. MATERIALS AND METHODS: Plasmids encoding the metabolic biosensors HyPer, iNap, Peredox, and Perceval were transfected into 15 ovarian cancer cell lines to assay oxidative stress, NADPH/NADP+, NADH/NAD+, and ATP/ADP, respectively. Fluorescence readings were used to assay dynamic metabolic responses to omental conditioned media (OCM) and 100 µM carboplatin treatment. SKOV3 cells expressing HyPer were imaged as 2D monolayers, 3D organoids, and as in vivo metastases via an intravital omental window. We further established organoids from ascites collected from Stage III/IV OC patients with carboplatin-resistant or carboplatin-sensitive tumors (n = 8 total). These patient-derived organoids (PDOs) were engineered to express HyPer, and metabolic readings of oxidative stress were performed during treatment with 100 µM carboplatin. RESULTS: Exposure to OCM or carboplatin induced heterogenous metabolic changes in 15 OC cell lines, as measured using metabolic sensors. Oxidative stress of in vivo omental metastases, measured via intravital imaging of metastasizing SKOV3-HyPer cells, was more closely recapitulated by SKOV3-HyPer organoids than by 2D monolayers. Finally, carboplatin treatment of HyPer-expressing PDOs induced higher oxidative stress in organoids derived from carboplatin-resistant patients than from those derived from carboplatin-sensitive patients. CONCLUSIONS: Our study showed that biosensors provide a useful method of studying dynamic metabolic changes in preclinical models of OC, including 3D organoids and intravital imaging. As 3D models of OC continue to evolve, the repertoire of biosensors will likely serve as valuable tools to probe the metabolic changes of clinical importance in OC.


Assuntos
Técnicas Biossensoriais , Neoplasias Ovarianas , Humanos , Feminino , Carboplatina/uso terapêutico , Carcinoma Epitelial do Ovário , NADP/uso terapêutico , NAD/uso terapêutico , Meios de Cultivo Condicionados , Neoplasias Ovarianas/metabolismo , Difosfato de Adenosina/uso terapêutico , Trifosfato de Adenosina/uso terapêutico , Microambiente Tumoral
4.
Stem Cell Reports ; 17(9): 1959-1975, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-35985334

RESUMO

In vitro tissue models hold great promise for modeling diseases and drug responses. Here, we used emulsion microfluidics to form micro-organospheres (MOSs), which are droplet-encapsulated miniature three-dimensional (3D) tissue models that can be established rapidly from patient tissues or cells. MOSs retain key biological features and responses to chemo-, targeted, and radiation therapies compared with organoids. The small size and large surface-to-volume ratio of MOSs enable various applications including quantitative assessment of nutrient dependence, pathogen-host interaction for anti-viral drug screening, and a rapid potency assay for chimeric antigen receptor (CAR)-T therapy. An automated MOS imaging pipeline combined with machine learning overcomes plating variation, distinguishes tumorspheres from stroma, differentiates cytostatic versus cytotoxic drug effects, and captures resistant clones and heterogeneity in drug response. This pipeline is capable of robust assessments of drug response at individual-tumorsphere resolution and provides a rapid and high-throughput therapeutic profiling platform for precision medicine.


Assuntos
Antineoplásicos , Organoides , Antineoplásicos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Microfluídica , Medicina de Precisão
5.
Cell Death Differ ; 28(8): 2351-2366, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33664479

RESUMO

Proliferative vitreoretinopathy (PVR) is a disease that causes severe blindness and is characterized by the formation of contractile fibrotic subretinal or epiretinal membranes. The epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is a hallmark of PVR. This work aims to examine the role of a long noncoding RNA (lncRNA) named EMT-related lncRNA in RPE (ERLR, LINC01705-201 (ENST00000438158.1)) in PVR and to explore the underlying mechanisms. In this study, we found that ERLR is upregulated in RPE cells stimulated with transforming growth factor (TGF)-ß1 as detected by lncRNA microarray and RT-PCR. Further studies characterized full-length ERLR and confirmed that it is mainly expressed in the cytoplasm. In vitro, silencing ERLR in RPE cells attenuated TGF-ß1-induced EMT, whereas overexpressing ERLR directly triggered EMT in RPE cells. In vivo, inhibiting ERLR in RPE cells reduced the ability of cells to induce experimental PVR. Mechanistically, chromatin immunoprecipitation (ChIP) assays indicated that the transcription factor TCF4 directly binds to the promoter region of ERLR and promotes its transcription. ERLR mediates EMT by directly binding to MYH9 protein and increasing its stability. TCF4 and MYH9 also mediate TGF-ß1-induced EMT in RPE cells. Furthermore, ERLR is also significantly increased in RPE cells incubated with vitreous PVR samples. In clinical samples of PVR membranes, ERLR was detected through fluorescent in situ hybridization (FISH) and colocalized with the RPE marker pancytokeratin (pan-CK). These results indicated that lncRNA ERLR is involved in TGF-ß1-induced EMT of human RPE cells and that it is involved in PVR. This finding provides new insights into the mechanism and treatment of PVR.


Assuntos
Células Epiteliais/metabolismo , RNA Longo não Codificante/genética , Epitélio Pigmentado da Retina/metabolismo , Vitreorretinopatia Proliferativa/metabolismo , Animais , Transição Epitelial-Mesenquimal , Humanos , Camundongos , Coelhos
6.
J Cell Mol Med ; 24(22): 13324-13335, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33047885

RESUMO

Exosomes have recently emerged as a pivotal mediator of many physiological and pathological processes. However, the role of exosomes in proliferative vitreoretinopathy (PVR) has not been reported. In this study, we aimed to investigate the role of exosomes in PVR. Transforming growth factor beta 2 (TGFß-2) was used to induce epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells, as an in vitro model of PVR. Exosomes from normal and EMTed RPE cells were extracted and identified. We incubated extracted exosomes with recipient RPE cells, and co-cultured EMTed RPE cells and recipient RPE cells in the presence of the exosome inhibitor GW4869. Both experiments suggested that there are further EMT-promoting effects of exosomes from EMTed RPE cells. MicroRNA sequencing was also performed to identify the miRNA profiles in exosomes from both groups. We identified 34 differentially expressed exosomal miRNAs (P <. 05). Importantly, miR-543 was found in exosomes from EMTed RPE cells, and miR-543-enriched exosomes significantly induced the EMT of recipient RPE cells. Our study demonstrates that exosomal miRNA is differentially expressed in RPE cells during EMT and that these exosomal miRNAs may play pivotal roles in EMT induction. Our results highlight the importance of exosomes as cellular communicators within the microenvironment of PVR.


Assuntos
Transição Epitelial-Mesenquimal , Exossomos/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Vitreorretinopatia Proliferativa/metabolismo , Compostos de Anilina/farmacologia , Compostos de Benzilideno/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Biblioteca Gênica , Humanos , MicroRNAs/metabolismo , Nanopartículas , Fator de Crescimento Transformador beta2/metabolismo
7.
Invest Ophthalmol Vis Sci ; 60(15): 5104-5111, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31826237

RESUMO

Purpose: Cell-cell contact in retinal pigment epithelium (RPE) involves adherent junctions, gap junctions, and tight junctions, which are primarily composed by E-cadherin, zona occludens 1 (ZO-1), and connexin 43, respectively. Here, we aimed to explore the relationship and interplay between these junction-associated proteins. Methods: E-cadherin, connexin 43, and ZO-1 expression in human primary RPE in the early phase after TGF-ß1 stimulation was detected. The knockdown of E-cadherin, ZO-1, and connexin 43 was performed to characterize the regulatory network involving these three proteins. Dye transfer and FITC-dextran permeability assays were conducted to observe the epithelial functional alterations. Transmission electron microscopy (TEM) was used to observe the ultrastructure of the cell-cell junctions in mouse RPE. The immunofluorescence staining and coimmunoprecipitation were performed to observe the colocalization and the physical association of E-cadherin, ZO-1, and connexin 43. Results: Among these three components, E-cadherin appeared to be the first protein that was downregulated after TGF-ß1 treatment. The ultrastructures of adherent junctions, gap junctions, and tight junctions could be observed in mouse RPE by TEM. E-cadherin, ZO-1, and connexin 43 were colocalized and physically bound to each other. The knockdown of one of these three proteins led to downregulation of the other two proteins and compromised epithelial function. Conclusions: E-cadherin, ZO-1, and connexin 43 were physically associated with each other and were mutually regulated. To enhance the understanding of cell-cell contacts, a holistic view is needed. Our results provide new insights in RPE disorders such as proliferative vitreoretinopathy.


Assuntos
Caderinas/genética , Conexina 43/genética , Regulação da Expressão Gênica , Epitélio Pigmentado da Retina/metabolismo , Vitreorretinopatia Proliferativa/genética , Proteína da Zônula de Oclusão-1/genética , Animais , Caderinas/biossíntese , Células Cultivadas , Conexina 43/biossíntese , Humanos , Junções Intercelulares , Camundongos , Microscopia Eletrônica de Transmissão , RNA/genética , Epitélio Pigmentado da Retina/ultraestrutura , Junções Íntimas , Vitreorretinopatia Proliferativa/metabolismo , Vitreorretinopatia Proliferativa/patologia , Proteína da Zônula de Oclusão-1/biossíntese
8.
FASEB J ; 33(3): 3212-3224, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30383450

RESUMO

The major pathogenesis of proliferative vitreoretinopathy (PVR) is that retinal pigment epithelial (RPE) cells undergo epithelial-mesenchymal transition (EMT) because of disordered growth factors, such as TGF-ß, in the vitreous humor. Bone morphogenetic proteins (BMPs) are pluripotent growth factors. In this study, we identified the antifibrotic activity of BMP7 in a PVR model both in vivo and in vitro. BMP7 expression was confirmed on the PVR proliferative membranes. BMP7 was down-regulated in the PVR vitreous humor and TGF-ß-induced RPE cell EMT. In the in vivo studies, BMP7 injection attenuated PVR progression in the eyes of the rabbit model. Additionally, BMP7 treatment maintained RPE cell phenotypes and relieved TGF-ß2-induced EMT, migration, and gel contraction in vitro. BMP7 inhibited the TGF-ß2-induced up-regulation of fibronectin and α-smooth muscle actin and the down-regulation of E-cadherin and zona occludens-1 by balancing the TGF-ß2/Smad2/3 and BMP7/Smad1/5/9 pathways. These findings provide direct evidence of the ability of BMP7 in PVR inhibition and the potential of BMP7 for use in PVR therapeutic intervention.-Yao, H., Ge, T., Zhang, Y., Li, M., Yang, S., Li, H., Wang, F. BMP7 antagonizes proliferative vitreoretinopathy through retinal pigment epithelial fibrosis in vivo and in vitro.


Assuntos
Proteína Morfogenética Óssea 7/metabolismo , Vitreorretinopatia Proliferativa/metabolismo , Animais , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/farmacologia , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Transição Epitelial-Mesenquimal , Fibrose , Técnicas de Silenciamento de Genes , Humanos , RNA Interferente Pequeno/genética , Coelhos , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta2/metabolismo , Vitreorretinopatia Proliferativa/patologia , Vitreorretinopatia Proliferativa/prevenção & controle , Corpo Vítreo/metabolismo
9.
Cell Physiol Biochem ; 46(4): 1704-1715, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29698960

RESUMO

BACKGROUND/AIMS: Proliferative vitreoretinopathy (PVR) is a severe blinding complication of rhegmatogenous retinal detachment. Epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is thought to play a pivotal role in the pathogenesis of PVR. Fucoidan, a marine extract, reportedly has many benefits effects in a variety of tissues and organs such as anti-inflammation, anti-oxidative stress, and anti-carcinogenesis. In this study, we investigated the potential role of fucoidan on EMT in RPE cells and its effect on the development of PVR. METHODS: MTS, Transwell, and collagen gel contraction assays were employed to measure the viability, migration, and contraction of RPE cells, respectively. mRNA and protein expression were evaluated via real-time quantitative PCR and western blot analysis, respectively. In vivo, a pigmented rabbit model of PVR was established to examine the anti-PVR effect of fucoidan. RESULTS: Fucoidan reversed the transforming growth factor (TGF)-ß1-induced EMT of RPE cells, including the increased expression of α-smooth muscle actin (α-SMA) and fibronectin and down-regulation of E-cadherin in human primary RPE cells. Moreover, the upregulation of phosphorylated Smad2/3 induced by TGF-ß1 was suppressed by fucoidan. Fucoidan also inhibited the migration and contraction of RPE cells induced by TGF-ß1. In vivo, fucoidan inhibited the progression of experimental PVR in rabbit eyes. Histological findings showed that fucoidan suppressed the formation of α-SMA-positive epiretinal membranes. CONCLUSION: Our findings regarding the protective effects of fucoidan on the EMT of RPE cells and experimental PVR suggest the potential clinical application of fucoidan as an anti-PVR agent.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Isoenzimas/metabolismo , Polissacarídeos/farmacologia , Substâncias Protetoras/farmacologia , Retinal Desidrogenase/metabolismo , Vitreorretinopatia Proliferativa/patologia , Actinas/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Caderinas/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Olho/diagnóstico por imagem , Fibronectinas/metabolismo , Humanos , Isoenzimas/genética , Estadiamento de Neoplasias , Fosforilação/efeitos dos fármacos , Polissacarídeos/uso terapêutico , Substâncias Protetoras/uso terapêutico , Coelhos , Retinal Desidrogenase/genética , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Tomografia de Coerência Óptica , Fator de Crescimento Transformador beta1/farmacologia , Vitreorretinopatia Proliferativa/tratamento farmacológico , Vitreorretinopatia Proliferativa/mortalidade
10.
Sci Rep ; 6: 32182, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27586653

RESUMO

Proliferative vitreoretinopathy (PVR), a serious vision-threatening complication of retinal detachment (RD), is characterized by the formation of contractile fibrotic membranes, in which epithelial-mesenchymal transition (EMT) of the retinal pigment epithelium (RPE) is a major event. Recent studies suggest an important role of bone morphogenetic protein 4 (BMP4) in the suppression of fibrosis. In this study, we aimed to investigate the role of BMP4 in the pathological process of PVR, particularly in the EMT of RPE cells. We found that BMP4 and its receptors were co-labelled with cytokeratin and α-SMA positive cells within the PVR membrane. Moreover, the mRNA and protein expression levels of BMP4 were decreased whereas BMP4 receptors ALK2, ALK3 and ALK6 were increased during TGF-ß-induced EMT in primary RPE cells. Exogenous BMP4 inhibited TGF-ß-induced epithelial marker down-regulation, as well as mesenchymal marker up-regulation at both the mRNA and protein levels in RPE cells. In addition, BMP4 treatment attenuated the TGF-ß-induced gel contraction, cell migration and Smad2/3 phosphorylation. However, knockdown of endogenous BMP4 stimulated changes in EMT markers. Our results confirm the hypothesis that BMP4 might inhibit TGF-ß-mediated EMT in RPE cells via the Smad2/3 pathway and suppress contraction. This might represent a potential treatment for PVR.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Epitélio Pigmentado da Retina/patologia , Vitreorretinopatia Proliferativa/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Movimento Celular/efeitos dos fármacos , Colágeno/metabolismo , Regulação para Baixo , Técnicas de Silenciamento de Genes , Humanos , Fosforilação/efeitos dos fármacos , Epitélio Pigmentado da Retina/efeitos dos fármacos , Proteína Smad1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Vitreorretinopatia Proliferativa/patologia
11.
PLoS One ; 11(3): e0152687, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27019196

RESUMO

PURPOSE: To study the role of long non-coding RNA (lncRNA) MALAT1 in transforming growth factor beta 1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells. METHODS: ARPE-19 cells were cultured and exposed to TGF-ß1. The EMT of APRE-19 cells is confirmed by morphological change, as well as the increased expression of alpha-smooth muscle actin (αSMA) and fibronectin, and the down-regulation of E-cadherin and Zona occludin-1(ZO-1) at both mRNA and protein levels. The expression of lncRNA MALAT1 in RPE cells were detected by quantitative real-time PCR. Knockdown of MALAT1 was achieved by transfecting a small interfering RNA (SiRNA). The effect of inhibition of MALAT1 on EMT, migration, proliferation, and TGFß signalings were observed. MALAT1 expression was also detected in primary RPE cells incubated with proliferative vitreoretinopathy (PVR) vitreous samples. RESULTS: The expression of MALAT1 is significantly increased in RPE cells incubated with TGFß1. MALAT1 silencing attenuates TGFß1-induced EMT, migration, and proliferation of RPE cells, at least partially through activating Smad2/3 signaling. MALAT1 is also significantly increased in primary RPE cells incubated with PVR vitreous samples. CONCLUSION: LncRNA MALAT1 is involved in TGFß1-induced EMT of human RPE cells and provides new understandings for the pathogenesis of PVR.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , RNA Longo não Codificante/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Actinas/metabolismo , Caderinas/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Fibronectinas/metabolismo , Humanos , Interferência de RNA , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Vitreorretinopatia Proliferativa/metabolismo , Vitreorretinopatia Proliferativa/patologia , Proteína da Zônula de Oclusão-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA