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.
Invest Ophthalmol Vis Sci ; 64(3): 35, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-37000125

RESUMO

Purpose: To explore the anti-inflammatory and neuroprotective effects of lithium chloride (LiCl) in LPS-induced retinal injury. Methods: In vitro, primary retinal microglia were pretreated with LiCl and stimulated with lipopolysaccharide (LPS). Pro-inflammatory cytokine production, microglial morphological changes, and inflammation-associated signaling pathways were measured by real-time PCR (RT-PCR), western blotting, and immunofluorescence. Primary retinal neurons were cultured with microglial-derived conditioned medium in the absence or presence of LiCl. Neurotoxicity was evaluated by Cell Counting Kit-8 (CCK-8), terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay, and γ-H2AX detection. In vivo, an endotoxin-induced uveitis mice model was established, and each animal was given intraperitoneal injection of LiCl or vehicle. The retinal inflammatory response was measured by hematoxylin and eosin and fluorescent staining, RT-PCR, western blotting, and TUNEL assay. Retinal thickness and function were evaluated by spectral-domain optical coherence tomography and electroretinography. Results: In vitro, LiCl exerted no obvious toxic effects on microglia and significantly decreased proinflammatory factor (inducible nitric oxide synthase, tumor necrosis factor α, interleukin 6) production, inhibited microglial activation in morphology, and suppressed nuclear factor kappa B (NF-κB), Akt, and phosphatidylinositol 3-kinase (PI3K) phosphorylation. Moreover, LiCl promoted retinal neuron survival and reduced cell apoptosis and the expression of γ-H2AX. In vivo, LiCl reduced inflammatory infiltrating cells in the vitreous cavity and decreased proinflammatory cytokine expression in retinas. LiCl suppressed LPS-induced microglial activation, proliferation, and migration. Additionally, LiCl reduced LPS-induced apoptosis of ganglion cells and retinal edema and rescued retinal functional damage. Conclusions: This study demonstrates that LiCl exerts anti-inflammatory and neuroprotective effects by inhibiting microglial activation via the PI3K/Akt/NF-κB pathway in LPS-induced retinal injury. LiCl provides a novel and promising option to treat retinal inflammatory diseases.


Assuntos
Fármacos Neuroprotetores , Doenças Retinianas , Camundongos , Animais , Lipopolissacarídeos/toxicidade , NF-kappa B/metabolismo , Microglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fármacos Neuroprotetores/metabolismo , Cloreto de Lítio/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Linhagem Celular , Anti-Inflamatórios/farmacologia , Doenças Retinianas/patologia , Citocinas/genética , Citocinas/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo
2.
Exp Eye Res ; 216: 108955, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35074341

RESUMO

Krüppel-like factor 2 (KLF2) belongs to the KLF family of zinc-finger transcription factors and mediates the occurrence and progression of various cancers. However, little is known about its expression pattern and biological role in retinoblastoma (RB). In the present study, we showed that KLF2 was markedly downregulated in human RB tissue compared with retina. KLF2 overexpression significantly inhibited RB cell proliferation and decreased proliferating cell nuclear antigen (PCNA) expression. Subsequently, we confirmed that KLF2 arrested cells at the G1-S phase transition, accompanied by the upregulation of p21 and downregulation of CyclinD1, as well as the activation of mitochondria-mediated apoptosis in RB cells. In addition, KLF2 overexpression contributed to suppressing RB cell migration and invasion by downregulating matrix metallopeptidase 9 (MMP9). On the contrary, KLF2 downregulation promoted RB cells proliferation, migration and invasion. Notably, the KLF2 expression pattern was opposite to that of C-X-C chemokine receptor 4 (CXCR4) in the two RB cell lines, KLF2 overexpression significantly decreased CXCR4 expression, silencing KLF2 had the opposite effect. Furthermore, dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays confirmed that KLF2 directly bound to the CXCR4 promoter and negatively regulated its expression in RB cells. Collectively, our results suggested that KLF2 function as a tumor suppressor in RB and may represent a potential therapeutic target for RB.


Assuntos
Fatores de Transcrição Kruppel-Like/fisiologia , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Apoptose/fisiologia , Western Blotting , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Marcação In Situ das Extremidades Cortadas , Plasmídeos , Antígeno Nuclear de Célula em Proliferação/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias da Retina/patologia , Retinoblastoma/patologia , Transfecção , Quinases Ativadas por p21/genética
3.
Cell Death Dis ; 12(7): 695, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34257272

RESUMO

Exosomes derived from tumor cells play a key role in tumor development. In the present study, we identified the bioactivity of exosomes released from WERI-Rb1 retinoblastoma cells in tumor angiogenesis, as well as the underlying mechanism, through biochemical methods and animal experiments. Our in vitro data showed that exosomes could be engulfed by human vesicle endothelial cells (HUVECs), significantly promote cell viability and induce an inflammatory response in HUVECs by increasing the expression of a series of related genes, such as IL-1, IL-6, IL-8, MCP-1, VCAM1, and ICAM1. Significant increases in migration and tube formation were also observed in the HUVECs incubated with exosomes. Moreover, experiments with a nude mouse xenotransplantation model showed that exosomes injected near tumors could be strongly absorbed by tumor cells. The numbers of endothelial cells and blood vessels were significantly increased in tumor tissues treated with exosomes compared to control tissues. Furthermore, to reveal the mechanism underlying exosome-mediated angiogenesis in retinoblastoma, we analyzed the levels of 12 microRNAs in the exosomes. Specifically, our data showed that miR-92a-3p was enriched in RB exosomes. Accordingly, miR-92a-3p was increased in the HUVECs incubated with these exosomes. After treatment with a miR-92a-3p inhibitor, the promoting effect of exosomes on the migration and tube formation of HUVECs was significantly abrogated. The expression of the angiogenesis-related genes mentioned above was markedly decreased in HUVECs. Similarly, treatment with a microRNA mimic also demonstrated that miR-92a-3p was involved in the angiogenesis of HUVECs. More importantly, bioinformatics analysis predicted that Krüppel-like factor 2 (KLF2), a member of the KLF family of zinc-finger transcription factors, might be an active target of miR-92a-3p. Notably, this prediction was confirmed both in vitro and in vivo. Thus, our work suggests that exosomal miR-92a-3p is involved in tumor angiogenesis and might be a promising therapeutic candidate for retinoblastoma.


Assuntos
Exossomos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , MicroRNAs/metabolismo , Neovascularização Patológica , Neovascularização Fisiológica , Comunicação Parácrina , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Exossomos/genética , Exossomos/patologia , Exossomos/transplante , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos Nus , MicroRNAs/genética , Neoplasias da Retina/genética , Neoplasias da Retina/patologia , Retinoblastoma/genética , Retinoblastoma/patologia
4.
Oncol Rep ; 45(1): 278-290, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33416154

RESUMO

The survival of young children (under 5 years of age) with malignant retinoblastoma remains poor, and clarification of the mechanism underlying tumour development is urgently needed. The present study aimed to reveal the role of exosomes (EXOs) from retinoblastoma cells in tumour development. The in vitro data indicated that EXOs derived from WERI­Rb1 cells significantly inhibited the antitumour activity of macrophages and induced bone marrow mesenchymal stem cells to promote tumour growth via an increase in monocyte chemotactic protein 1 (also known as C­C motif chemokine ligand 2) levels. In vivo data from a xenotransplantation model also showed that EXOs infiltrated the spleen, which induced a decrease in leukocytes and natural killer (NK) cells. Accordingly, the proportion of tumour­associated macrophages was increased and the proportion of NK cells was decreased in tumours injected with EXOs compared with those injected with the control. EXOs were absorbed by Kupffer cells, and more metastases were observed in the liver. Thus, these results suggested that EXOs derived from retinoblastoma promoted tumour progression by infiltrating the microenvironment. Moreover, microRNAs (miRs), including miR­92a, miR­20a, miR­129a and miR­17, and C­X­C chemokine receptor type 4 and thrombospondin­1 were detectable in EXOs, which might account for EXO­mediated tumour deterioration.


Assuntos
Carcinogênese/imunologia , Exossomos/imunologia , Retinoblastoma/imunologia , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/imunologia , Animais , Carcinogênese/patologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Quimiocina CCL2/metabolismo , Pré-Escolar , Técnicas de Cocultura , Exossomos/metabolismo , Feminino , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Células-Tronco Mesenquimais , Camundongos , MicroRNAs/metabolismo , Cultura Primária de Células , Retinoblastoma/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biomed Pharmacother ; 128: 110273, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32460188

RESUMO

Ocular inflammation is a common pathological condition of a series of retinal degenerative diseases. Tetramethylpyrazine (TMP), a Chinese herbal extraction, is widely used in the treatment of several ocular diseases in Eastern countries. However, the exact mechanisms correlating the vision protective effects of TMP have not been elucidated. Thus, this study aimed to investigate TMP's molecular targets in anti-inflammatory activity in endotoxin lipopolysaccharide (LPS)-induced retinal inflammation both in vitro and in vivo. The primary cultured retinal microglial cells were pretreated with TMP and then activated by LPS. We found pretreatment with TMP significantly inhibited LPS-induced upregulation of CD68, a marker of mononuclear microglia activation. The morphological changes induced by LPS were also inhibited by the TMP pretreatment. Moreover, Toll like receptor 4 (TLR4), phosphorylation of inhibitor of NF-κB alpha (p-IκB-α) and the translocation of nuclear factor kappa B p65 (NF-κB p65) were significantly downregulated in retinal microglial cells with TMP pretreatment, which indicated that TMP might suppress LPS-induced retinal microglial activation through TLR4/NF-κB signalling pathway. And these results were confirmed in vivo. Pretreatment with TMP inhibited microglial activation, migration and regeneration, especially in ganglion cell layer (GCL). In addition to the inhibition of TLR4, TMP significantly inhibited the translocation of NF-κB p-65 to the nucleus in vivo. The downstream genes of NF-κB, such as the pro-inflammatory cytokines interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α) and interleukin-1ß (IL-1ß), were significantly downregulated by TMP pretreatment in the retina. Accordingly, the increased expression of cleaved caspase-3 and the decreased ratio of B-cell lymphoma-2 (Bcl-2) to Bcl-2 associated X Protein (Bax) were significantly attenuated by TMP. TUNEL assay also demonstrated that TMP exerted neuroprotective effects in the retina. Therefore, this study elucidated a novel mechanism that TMP inhibits retinal inflammation by inhibiting microglial activation via a TLR4/NF-κB signalling pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Microglia/efeitos dos fármacos , NF-kappa B/metabolismo , Pirazinas/farmacologia , Células Ganglionares da Retina/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Uveíte/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Microglia/metabolismo , Microglia/patologia , Ratos Sprague-Dawley , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Transdução de Sinais , Uveíte/induzido quimicamente , Uveíte/metabolismo , Uveíte/patologia
6.
Cell Death Dis ; 10(11): 862, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31723124

RESUMO

Retinoblastoma (RB) is the most common primary intraocular malignancy in children. Intravitreal chemotherapy achieves favorable clinical outcomes in controlling RB vitreous seeds, which are a common reason for treatment failure. Thus, a novel, effective and safe intravitreal chemotherapeutic drug is urgently required. The malaria drug artesunate (ART) recently demonstrated remarkable anticancer effects with mild side effects. The purpose of this study is to investigate the anti-RB efficacy, the underlying mechanism and the intraocular safety of ART. Herein, we verified that ART inhibits RB cell viability and induces cell apoptosis in a dose- and time-dependent manner. Microarray analysis revealed that Kruppel-like factor 6 (KLF6) was upregulated after ART treatment, and this was further confirmed by real-time PCR and western blot assays. Silencing of KLF6 expression significantly reversed ART-induced RB cell growth inhibition and apoptosis. Furthermore, ART activated mitochondria-mediated apoptosis of RB cells, while silencing KLF6 expression significantly inhibited this effect. In murine xenotransplantation models of RB, we further confirmed that ART inhibits RB tumor growth, induces tumor cell apoptosis and upregulates KLF6 expression. In addition, KLF6 silencing attenuates ART-mediated inhibition of tumor growth in vivo. Furthermore, we proved that intravitreal injection of ART in Sprague-Dawley (SD) rats is safe, with no obvious retinal function damage or structural disorders observed by electrophysiology (ERG), fundal photographs, fundus fluorescein angiography (FFA) or optical coherence tomography (OCT) examinations. Collectively, our study revealed that ART induces mitochondrial apoptosis of RB cells via upregulating KLF6, and our results may extend the application of ART to the clinic as an effective and safe intravitreal chemotherapeutic drug to treat RB, especially RB with vitreous seeds.


Assuntos
Artesunato/farmacologia , Proliferação de Células/efeitos dos fármacos , Fator 6 Semelhante a Kruppel/genética , Retinoblastoma/tratamento farmacológico , Animais , Antimaláricos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Reposicionamento de Medicamentos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Xenoenxertos , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Ratos , Retinoblastoma/genética , Retinoblastoma/patologia , Ativação Transcricional/efeitos dos fármacos
7.
Oncol Rep ; 42(3): 1214-1224, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31322174

RESUMO

Tetramethylpyrazine (TMP; an extract of the Chinese herbal medicine, Chuanxiong) has been shown to exert remarkable antiretinoblastoma (RB) effects. Based on our previous study, the target gene was found to be C­X­C chemokine receptor type 4 (CXCR4). CXCR4 is a prognostic marker in various types of cancer, but the exact mechanisms underlying the regulation of CXCR4 expression by TMP in WERI­Rb1 cells have yet to be fully elucidated. In the present study, it was revealed that TMP significantly downregulated CXCR4 expression and inhibited CXCR4 promoter activity in WERI­Rb1 cells, indicating that TMP inhibits CXCR4 expression in WERI­Rb1 cells through transcriptional regulatory mechanisms. Among the numerous transcription factors involved in CXCR4 function, including Yin Yang 1 (YY1), nuclear respiratory factor­1 (Nrf­1), Krüppel­like Factor 2 (KLF2), specificity protein 1 (SP1), and nuclear factor­κB subunit 1 (NF­κB1), only TMP led to a significant downregulation of Nrf­1 expression. Chromatin immunoprecipitation assays further indicated that Nrf­1 directly binds to the promoter region of CXCR4, and silencing Nrf­1 via siRNA transfection notably inhibited CXCR4 expression in WERI­Rb1 cells. In addition, the expression levels of both Nrf­1 and CXCR4 increased concomitantly with WERI­Rb1 cell density. Furthermore, the downregulation of Nrf­1 and CXCR4 expression in RB by TMP was confirmed in vivo. Taken together, the results of the present study have uncovered a novel mechanism in which CXCR4 expression is regulated by Nrf­1 in WERI­Rb1 cells, thereby identifying novel potential targets for the treatment of RB, and providing evidence for the clinical application of TMP in adjuvant retinoblastoma therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator 1 Nuclear Respiratório/metabolismo , Pirazinas/farmacologia , Receptores CXCR4/genética , Neoplasias da Retina/patologia , Retinoblastoma/patologia , Transcrição Gênica/efeitos dos fármacos , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Nus , Fator 1 Nuclear Respiratório/genética , Receptores CXCR4/metabolismo , Neoplasias da Retina/tratamento farmacológico , Neoplasias da Retina/genética , Retinoblastoma/tratamento farmacológico , Retinoblastoma/genética , Células Tumorais Cultivadas , Vasodilatadores/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Mol Med Rep ; 15(5): 2481-2488, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28447713

RESUMO

Retinoblastoma is the most common ocular tumor in children, and it causes extensive damage. Current treatment options for retinoblastoma include surgery, chemotherapy, radiotherapy and cryotherapy. However, the majority of chemotherapy medicines cause complications and side effects that lead to severe impairment of patient health. Previous studies have reported that tetramethylpyrazine (TMP), which is an extract of the Chinese herbal medicine Chuanxiong, reduces the risk of multidrug resistance in chemotherapy and inhibits the proliferation and metastasis of various types of cancer cells. However, the underlying molecular mechanism of TMP in retinoblastoma remains unclear. The current study demonstrated that C-X-C chemokine receptor type 4 (CXCR4) was expressed in WERI­Rb1 cells and in retinoblastoma. Using reverse transcription­quantitative polymerase chain reaction and western blotting techniques, the current study demonstrated that TMP significantly downregulated the expression of CXCR4 in WERI­Rb1 cells cultured at high density, whereas it had a minor effect in low­density WERI­Rb1 cells; additionally, this effect occurred in a time­dependent manner. TMP inhibited the proliferation of WERI­Rb1 cells as effectively as a CXCR4 antagonist, AMD3100, consistent with a role of CXCR4 in cancer development. Notably, TMP did not affect the cell cycle of cells cultured at low density (1x105 cells/ml), whereas it induced G1­phase arrest in high­density cells (7.5x105 cells/ml; P<0.05). In addition, the expression of CXCR4 in primary rat retinal neurocytes was significantly downregulated by TMP treatment, and this treatment protected primary rat retinal neurocytes from H2O2­induced damage. Thus, the results of this study indicate that TMP is a potential candidate for use in treatment of retinoblastoma, and also provides novel insights into the mechanisms of the anti­cancer and neuroprotective effects of this extract.


Assuntos
Proteínas de Neoplasias/biossíntese , Pirazinas/farmacologia , Receptores CXCR4/biossíntese , Retinoblastoma/metabolismo , Adolescente , Adulto , Animais , Linhagem Celular Tumoral , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ratos , Retinoblastoma/tratamento farmacológico , Retinoblastoma/patologia
9.
Biochem Biophys Res Commun ; 484(2): 285-291, 2017 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-28126341

RESUMO

Cobalt chloride (CoCl2) has long been accepted as a suitable in vitro hypoxia-mimetic agent. The gene CXCR4, which encodes a chemokine receptor, plays a key role in hypoxic retinal disease. Here, we investigated the mRNA and protein expression of CXCR4 in WERI-Rb1 retinoblastoma cells and human umbilical vein endothelial cells (HUVECs) under CoCl2-induced hypoxic conditions, by means of real-time PCR and western blot. We found that CoCl2-induced hypoxia profoundly increased CXCR4 expression at the mRNA level, but not at the protein level, at 12, 24, 48 and 72 h in these cells. Interestingly, this result differed from observations of 1% O2 hypoxic conditions. Additionally, luciferase assays demonstrated that CoCl2-induced hypoxia significantly increased transcription at the CXCR4 promoter. In order to compare our in vitro findings with the effects of hypoxia in vivo, an OIR (Oxygen-induced retinopathy) rat model was constructed. However, both CXCR4 mRNA and protein levels in OIR rats were significantly increased compared to controls. Thus taken together, our findings suggest that the relationship between CXCR4 mRNA and protein expression is not strictly linear under in vitro CoCl2-induced hypoxic conditions. through comparative in vitro and in vivo experiments, this study implies that CoCl2 is an imperfect simulation of hypoxia in retinal disease.


Assuntos
Hipóxia Celular , Cobalto/farmacologia , RNA Mensageiro/metabolismo , Receptores CXCR4/metabolismo , Animais , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana , Humanos , Ratos , Receptores CXCR4/genética , Doenças Retinianas/metabolismo , Regulação para Cima
10.
Cell Death Dis ; 7(11): e2473, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27853172

RESUMO

Neurons display genomic fragility and show fragmented DNA in pathological degeneration. A failure to repair DNA breaks may result in cell death or apoptosis. Lithium protects retinal neurocytes following nutrient deprivation or partial nerve crush, but the underlying mechanisms are not well defined. Here we demonstrate that pretreatment with lithium protects retinal neurocytes from ischemia-induced damage and enhances light response in rat retina following ischemia-reperfusion injury. Moreover, we found that DNA nonhomologous end-joining (NHEJ) repair is implicated in this process because in ischemic retinal neurocytes, lithium significantly reduces the number of γ-H2AX foci (well-characterized markers of DNA double-strand breaks in situ) and increases the DNA ligase IV expression level. Furthermore, we also demonstrate that nuclear respiratory factor 1 (Nrf-1) and phosphorylated cyclic AMP-response element binding protein-1 (P-CREB1) bind to ligase IV promoter to cause upregulation of ligase IV in neurocytes. The ischemic upregulation of Nrf-1 and lithium-induced increase of P-CREB1 cooperate to promote transcription of ligase IV. Short hairpin RNAs against Nrf-1 and CREB1 could significantly inhibit the increase in promoter activity and expression of ligase IV observed in the control oligos following lithium treatment in retinal neurocytes. More importantly, ischemic stimulation triggers the expression of ligase IV. Taken together, our results thus reveal a novel mechanism that lithium offers neuroprotection from ischemia-induced damage by enhancing DNA NHEJ repair.


Assuntos
DNA Ligase Dependente de ATP/metabolismo , DNA/metabolismo , Isquemia/patologia , Lítio/farmacologia , Neurônios Retinianos/enzimologia , Neurônios Retinianos/patologia , Regulação para Cima/efeitos dos fármacos , Animais , Sequência de Bases , Sítios de Ligação , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Imunoprecipitação da Cromatina , Meios de Cultura Livres de Soro , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dano ao DNA , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Modelos Animais de Doenças , Eletrorretinografia , Inativação Gênica , Isquemia/enzimologia , Luz , Fator 1 Relacionado a NF-E2/metabolismo , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/metabolismo , Ratos , Traumatismo por Reperfusão/patologia , Neurônios Retinianos/efeitos dos fármacos , Neurônios Retinianos/efeitos da radiação , Transcrição Gênica/efeitos dos fármacos
11.
Oncotarget ; 7(5): 6105-20, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26756218

RESUMO

Retinoblastoma (RB) arises from the retina, and its growth usually occurs under the retina and toward the vitreous. Ideal therapy should aim to inhibit the tumor and protect neural cells, increasing the patient's life span and quality of life. Previous studies have demonstrated that Thrombospondin-1 (TSP-1) is associated with neurogenesis, neovascularization and tumorigenesis. However, at present, the bioactivity of TSP-1 in retinoblastoma has not been defined. Herein, we demonstrated that TSP-1 was silenced in RB cell lines and clinical tumor samples. HDAC inhibitor, Trichostatin A (TSA), could notably transcriptionally up-regulate TSP-1 in RB cells, WERI-Rb1 cells and Y79 cells. Moreover, we found human recombinant TSP-1 (hTSP-1) could significantly inhibit the cell viability of RB cells both in vitro and in vivo. Interestingly, hTSP-1 could significantly induce the expression of γ-H2AX, a well-characterized in situ marker of DNA double-strand breaks (DSBs) in RB cells. The DNA NHEJ pathway in WERI-Rb1 cells could be significantly inhibited by hTSP-1. A mutation in Rb1 might be involved in the hTSP-1-medicated γ-H2AX increasing in WERI-Rb1 cells. Furthermore, hTSP-1 could inhibit RB cells while promoting retinal neurocyte survival in the neuronal and retinoblastoma cell co-culture system. As such, TSP-1 may become a therapeutic target for treatment of retinoblastoma.


Assuntos
Quebras de DNA de Cadeia Dupla , Histonas/metabolismo , Proteína do Retinoblastoma/metabolismo , Retinoblastoma/terapia , Trombospondina 1/administração & dosagem , Acetilação/efeitos dos fármacos , Animais , Histonas/genética , Humanos , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Distribuição Aleatória , Retinoblastoma/tratamento farmacológico , Retinoblastoma/genética , Retinoblastoma/metabolismo , Trombospondina 1/genética , Trombospondina 1/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...