Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 857
Filtrar
1.
Stem Cell Res ; 76: 103373, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38452707

RESUMO

Complete loss of RB1 causes retinoblastoma. Here, we report the generation of three RB1-/- iPSC lines using CRISPR/Cas9 based editing at exon 18 of RB1 in a healthy control hiPSC line. The edited cells were clonally expanded, genotyped and characterized to establish the mutant lines. Two of the mutant lines are compound heterozygous, with different in-del mutations in each of their alleles, while the third mutant line is homozygous, with identical edits in both alleles. All lines maintained their stemness, pluripotency, formed embryoid bodies with cell types of all three lineages, displayed a normal karyotype and lost RB1 expression.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias da Retina , Retinoblastoma , Humanos , Retinoblastoma/genética , Retinoblastoma/metabolismo , Sistemas CRISPR-Cas/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação , Neoplasias da Retina/genética , Neoplasias da Retina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Ligação a Retinoblastoma/genética
2.
Stem Cell Res ; 76: 103329, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38335663

RESUMO

Retinoblastoma is a pediatric intraocular cancer caused by biallelic inactivation of RB1 gene in retinal progenitor cells. Here, we report the generation of a patient-specific induced pluripotent stem cell (iPSC) line (LVPEIi002-A) from a patient diagnosed with retinoblastoma and showing familial inheritance of a nonsense mutation (c.1735C > T) within exon 18 of one of the two alleles. This RB1+/- iPSC line, LVPEIi002-A was generated by reprogramming the peri-orbital fat tissue derived mesenchymal cells and was stably expanded and characterized. It maintains the stemness, pluripotency, normal karyotype, and forms embryoid bodies comprising of all three lineage committed progenitor cells.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias da Retina , Retinoblastoma , Criança , Humanos , Retinoblastoma/genética , Retinoblastoma/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação/genética , Retina/metabolismo , Neoplasias da Retina/genética , Neoplasias da Retina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Ligação a Retinoblastoma/genética
3.
Int J Mol Sci ; 25(3)2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38339011

RESUMO

In childhood, retinoblastoma (RB) is the most common primary tumor in the eye. Long term therapeutic management with etoposide of this life-threatening condition may have diminishing effectiveness since RB cells can develop cytostatic resistance to this drug. To determine whether changes in receptor-mediated control of Ca2+ signaling are associated with resistance development, fluorescence calcium imaging, semi-quantitative RT-qPCR analyses, and trypan blue dye exclusion staining patterns are compared in WERI-ETOR (etoposide-insensitive) and WERI-Rb1 (etoposide-sensitive) cells. The cannabinoid receptor agonist 1 (CNR1) WIN55,212-2 (40 µM), or the transient receptor potential melastatin 8 (TRPM8) agonist icilin (40 µM) elicit similar large Ca2+ transients in both cell line types. On the other hand, NGF (100 ng/mL) induces larger rises in WERI-ETOR cells than in WERI-Rb1 cells, and its lethality is larger in WERI-Rb1 cells than in WERI-ETOR cells. NGF and WIN55,212-2 induced additive Ca2+ transients in both cell types. However, following pretreatment with both NGF and WIN55,212-2, TRPM8 gene expression declines and icilin-induced Ca2+ transients are completely blocked only in WERI-ETOR cells. Furthermore, CNR1 gene expression levels are larger in WERI-ETOR cells than those in WERI-Rb1 cells. Therefore, the development of etoposide insensitivity may be associated with rises in CNR1 gene expression, which in turn suppress TRPM8 gene expression through crosstalk.


Assuntos
Receptor de Fator de Crescimento Neural , Neoplasias da Retina , Retinoblastoma , Canais de Cátion TRPM , Humanos , Linhagem Celular , Etoposídeo/farmacologia , Etoposídeo/uso terapêutico , Proteínas de Membrana/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Neoplasias da Retina/tratamento farmacológico , Retinoblastoma/tratamento farmacológico , Retinoblastoma/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Receptor CB1 de Canabinoide/metabolismo
4.
Glia ; 72(5): 872-884, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38258347

RESUMO

RB1 deficiency leads to retinoblastoma (Rb), the most prevalent intraocular malignancy. Tumor-associated macrophages (TAMs) are related to local inflammation disorder, particularly by increasing cytokines and immune escape. Microglia, the unique resident macrophages for retinal homeostasis, are the most important immune cells of Rb. However, whether RB1 deficiency affects microglial function remain unknown. In this study, microglia were successfully differentiated from Rb patient- derived human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs), and then we investigated the function of RB1 in microglia by live imaging phagocytosis assay, immunofluorescence, RNA-seq, qRT-PCR, ELISA and retina organoids/microglia co-culturing. RB1 was abundantly expressed in microglia and predominantly located in the nucleus. We then examined the phagocytosis ability and secretion function of iMGs in vitro. We found that RB1 deficiency did not affect the expression of microglia-specific markers or the phagocytic abilities of these cells by live-imaging. Upon LPS stimulation, RB1-deficient microglia displayed enhanced innate immune responses, as evidenced by activated MAPK signaling pathway and elevated expression of IL-6 and TNF-α at both mRNA and protein levels, compared to wildtype microglia. Furthermore, retinal structure disruption was observed when retinal organoids were co-cultured with RB1-deficient microglia, highlighting the potential contribution of microglia to Rb development and potential therapeutic strategies for retinoblastoma.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias da Retina , Retinoblastoma , Humanos , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Microglia/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Retina , Neoplasias da Retina/genética , Neoplasias da Retina/metabolismo , Neoplasias da Retina/patologia
5.
Commun Biol ; 7(1): 11, 2024 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-38172218

RESUMO

Retinoblastoma (RB) is the most prevalent ocular tumor of childhood, and its extraocular invasion significantly increases the risk of metastasis. Nevertheless, a single-cell characterization of RB local extension has been lacking. Here, we perform single-cell RNA sequencing on four RB samples (two from intraocular and two from extraocular RB patients), and integrate public datasets of five normal retina samples, four intraocular samples, and three extraocular RB samples to characterize RB local extension at the single-cell level. A total of 128,454 qualified cells are obtained in nine major cell types. Copy number variation inference reveals chromosome 6p amplification in cells derived from extraocular RB samples. In cellular heterogeneity analysis, we identified 10, 8, and 7 cell subpopulations in cone precursor like cells, retinoma like cells, and MKI67+ photoreceptorness decreased (MKI67+ PhrD) cells, respectively. A high expression level of SOX4 was detected in cells from extraocular samples, especially in MKI67+ PhrD cells, which was verified in additional clinical RB samples. These results suggest that SOX4 might drive RB local extension. Our study presents a single-cell transcriptomic landscape of intraocular and extraocular RB samples, improving our understanding of RB local extension at the single-cell resolution and providing potential therapeutic targets for RB patients.


Assuntos
Neoplasias da Retina , Retinoblastoma , Humanos , Retinoblastoma/metabolismo , Variações do Número de Cópias de DNA , Neoplasias da Retina/genética , Neoplasias da Retina/patologia , Perfilação da Expressão Gênica , Fatores de Transcrição SOXC/genética
6.
Curr Med Sci ; 44(1): 223-231, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38277016

RESUMO

OBJECTIVE: Retinoblastoma (RB) is a prevalent type of eye cancer in youngsters. Prospero homeobox 1 (Prox1) is a homeobox transcriptional repressor and downstream target of the proneural gene that is relevant in lymphatic, hepatocyte, pancreatic, heart, lens, retinal, and cancer cells. The goal of this study was to investigate the role of Prox1 in RB cell proliferation and drug resistance, as well as to explore the underlying Notch1 mechanism. METHODS: Human RB cell lines (SO-RB50 and Y79) and a primary human retinal microvascular endothelial cell line (ACBRI-181) were used in this study. The expression of Prox1 and Notch1 mRNA and protein in RB cells was detected using quantitative real time-polymerase chain reaction (RT-qPCR) and Western blotting. Cell proliferation was assessed after Prox1 overexpression using the Cell Counting Kit-8 and the MTS assay. Drug-resistant cell lines (SO-RB50/vincristine) were generated and treated with Prox1 to investigate the role of Prox1 in drug resistance. We employed pcDNA-Notch1 to overexpress Notch1 to confirm the role of Notch1 in the protective function of Prox1. Finally, a xenograft model was constructed to assess the effect of Prox1 on RB in vivo. RESULTS: Prox1 was significantly downregulated in RB cells. Overexpression of Prox1 effectively decreased RB cell growth while increasing the sensitivity of drug-resistant cells to vincristine. Notch1 was involved in Prox1's regulatory effects. Notch1 was identified as a target gene of Prox1, which was found to be upregulated in RB cells and repressed by increased Prox1 expression. When pcDNA-Notch1 was transfected, the effect of Prox1 overexpression on RB was removed. Furthermore, by downregulating Notch1, Prox1 overexpression slowed tumor development and increased vincristine sensitivity in vivo. CONCLUSION: These data show that Prox1 decreased RB cell proliferation and drug resistance by targeting Notch1, implying that Prox1 could be a potential therapeutic target for RB.


Assuntos
Neoplasias da Retina , Retinoblastoma , Humanos , Linhagem Celular Tumoral , Proliferação de Células/genética , Resistência a Medicamentos , Neoplasias da Retina/tratamento farmacológico , Neoplasias da Retina/genética , Neoplasias da Retina/metabolismo , Retinoblastoma/tratamento farmacológico , Retinoblastoma/genética , Retinoblastoma/metabolismo , Vincristina/farmacologia
7.
Mol Biotechnol ; 66(1): 102-111, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37041423

RESUMO

Retinoblastoma (RB) is a malignant ocular cancer that affects children. Several microRNAs (miRNAs) have been implicated in RB regulation. The present study aimed to investigate the role of miR-4529-3p in RB pathogenesis. Scratch, Transwell, and Cell Counting Kit (CCK)-8 assays were conducted to assess the migratory, invasive, and proliferative abilities of RB cells. The expression levels of miR-4529-3p, RB1, and ERK pathway-related proteins were analyzed using western blotting and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR). Target relationships were verified using dual-luciferase reporter experiments. A murine RB model was developed to analyze the effects of miR-4529-3p on RB tumor growth in vivo. Our experiments revealed high levels of miR-4529-3p and low levels of RB1 in RB tissues. Functional analyses revealed that the migratory, invasive, and proliferative abilities of RB cells were repressed by miR-4529-3p inhibition. Similarly, p-ERK 1/2 protein levels were suppressed by miR-4529-3p inhibition. Furthermore, downregulation of miR-4529-3p limited tumor growth in vivo. Mechanistically, miR-4259-3p targets RB1. Interestingly, RB1 silencing abrogated the alleviative effects of miR-4529-3p downregulation in RB cells. MiR-4529-3p promotes RB progression by inhibiting RB1 and activating the ERK pathway. This evidence suggests that the miR-4529-3p/RB1 regulatory axis may be a prospective target for RB treatment in clinical settings.


Assuntos
MicroRNAs , Neoplasias da Retina , Retinoblastoma , Criança , Humanos , Animais , Camundongos , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Sistema de Sinalização das MAP Quinases/genética , Linhagem Celular Tumoral , MicroRNAs/genética , MicroRNAs/metabolismo , Transdução de Sinais , Neoplasias da Retina/genética , Neoplasias da Retina/metabolismo , Neoplasias da Retina/patologia , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Ligação a Retinoblastoma/genética , Proteínas de Ligação a Retinoblastoma/metabolismo
8.
Naunyn Schmiedebergs Arch Pharmacol ; 397(2): 1003-1013, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37555853

RESUMO

Retinoblastoma is a prevalent pediatric intraocular tumor. The suppressive effect of gentiopicroside (GPS) has been reported on various tumors. This study sought to determine the effect of GPS on retinoblastoma cell proliferation, apoptosis, invasion, and epithelial-mesenchymal transition (EMT), and tumorigenesis in nude mice. The effect and mechanism of GPS on growth, apoptosis, invasion, and EMT were determined by cell counting kit-8 (CCK-8), western blot, flow cytometry, and transwell assays in retinoblastoma cells. Y79 cells were injected into the vitreous cavity of BALB/c­nude mice to construct a retinoblastoma mouse model. Tumor growth and mouse weight were monitored for sequential 5 weeks. The effect of GPS in vivo was assessed by immunohistochemistry (IHC), terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling (TUNEL), and western blot assays. GPS decreased the cell viability of both Y79 and Weri-Rb1 cells with the IC50 of 18.85 µM and 27.57 µM, respectively. Besides, GPS reduced the relative expression of proteins involved in proliferation and EMT, and the number of invading cells, while increased the apoptosis rate and the relative expressions of apoptosis proteins in retinoblastoma cells. Mechanically, GPS decreased the relative protein level of PI3K/AKT pathway, which was then recovered after 740 Y-P was applied. Correspondingly, 740 Y-P reversed the inhibitory effect of GPS on growth, invasion, and EMT, and the increased effect of GPS on apoptosis. Additionally, GPS decreased tumor volume and weight as well as the relative level of Ki-67, VEGF, p-PI3K/PI3K, and p-AKT/AKT, while increased the apoptosis rate in vivo. GPS inhibited retinoblastoma cell proliferation and invasion via deactivating the PI3K/AKT pathway in both cell and animal models.


Assuntos
Glucosídeos Iridoides , Neoplasias da Retina , Retinoblastoma , Criança , Humanos , Animais , Camundongos , Retinoblastoma/tratamento farmacológico , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Proliferação de Células , Apoptose , Carcinogênese , Neoplasias da Retina/tratamento farmacológico , Neoplasias da Retina/metabolismo , Neoplasias da Retina/patologia , Movimento Celular
9.
Chin Clin Oncol ; 12(5): 52, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37964544

RESUMO

BACKGROUND: Retinoblastoma (RB) is a retinal cancer most commonly occurred in young children. Cisplatin and etoposide had been confirmed as chemotherapy drugs in the treatment of RB, even though the phenomenon of chemotherapeutic resistance has been occurring in clinical treatment frequently. RB has been reported to be a tumor with reduced expression of yes-associated protein (YAP). However, the role of YAP protein and its correlation with the chemotherapy effect in RB still remains unknown. METHODS: Here we used human RB cell lines Y79 and RB3823 to construct YAP over-expression cell lines for exploring the specific role of YAP. In vitro, a series of techniques and methods were used to identify the biological role of YAP in RB, such as Agilent Seahorse assay, lipid peroxidation assay, intracellular reactive oxygen species (ROS) measurement, flow cytometry apoptosis assay, and other basic experimental techniques, among others. RESULTS: The cell growth and cytology experimental results found YAP can inhibit the proliferation of RB cells and promote their apoptosis (Y79 32.71% vs. 3.75%; RB3823 40.32% vs. 6.73%). The mitochondrial fuel flex test, lipid peroxide and ROS measurement confirmed that YAP over-expression could promote mitochondrial fatty-acids ß-oxidation and lipid peroxidation in RB cells. Quantitative real-time polymerase chain reaction (qRT-PCR) analysis for the expression of lipid peroxidation related factors imply that YAP over-expression caused ferroptosis in RB cell lines. In addition, YAP transcription specific activator PY-60 (10 µM) further improved the sensitivity of cisplatin/etoposide. CONCLUSIONS: Our research results found the expression of YAP inhibits cell proliferation and promoted lipid peroxidation induced ferroptosis in RB. Interestingly, the mitochondrial oxidative phosphorylation shows an increased fatty acid dependency and decreased glucose dependency. As a result, this phenomenon improved the sensitivity of RB to cisplatin/etoposide chemotherapy in vitro. Our finding provides a potential therapeutic target for RB chemotherapy.


Assuntos
Ferroptose , MicroRNAs , Neoplasias da Retina , Retinoblastoma , Criança , Humanos , Pré-Escolar , Retinoblastoma/tratamento farmacológico , Retinoblastoma/genética , Retinoblastoma/metabolismo , Etoposídeo/farmacologia , Etoposídeo/uso terapêutico , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Peroxidação de Lipídeos , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Espécies Reativas de Oxigênio/uso terapêutico , Linhagem Celular Tumoral , Neoplasias da Retina/tratamento farmacológico , Neoplasias da Retina/genética , Neoplasias da Retina/patologia , Proliferação de Células , Lipídeos/farmacologia , Lipídeos/uso terapêutico , MicroRNAs/metabolismo , Regulação Neoplásica da Expressão Gênica
10.
Stem Cell Res ; 72: 103200, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37708614

RESUMO

Retinoblastoma (RB) is a common intraocular malignancy mostly caused by variation of the tumour suppressor gene RB1. In this study, we successfully generated two induced pluripotent stem cell (iPSC) lines from an infant with non-heritable RB. Both cell clones exhibited typical iPSC characteristics with normal karyotypes, consistent pluripotency markers expression and the capability of trilineage differentiation.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias da Retina , Retinoblastoma , Lactente , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Retinoblastoma/genética , Retinoblastoma/metabolismo , Diferenciação Celular/genética , Neoplasias da Retina/genética , Neoplasias da Retina/metabolismo , Neoplasias da Retina/patologia , Túnica Conjuntiva/metabolismo , Túnica Conjuntiva/patologia
11.
Transl Res ; 261: 41-56, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37419277

RESUMO

Lack of retinoblastoma (Rb) protein causes aggressive intraocular retinal tumors in children. Recently, Rb tumors have been shown to have a distinctly altered metabolic phenotype, such as reduced expression of glycolytic pathway proteins alongside altered pyruvate and fatty acid levels. In this study, we demonstrate that loss of hexokinase 1(HK1) in tumor cells rewires their metabolism allowing enhanced oxidative phosphorylation-dependent energy production. We show that rescuing HK1 or retinoblastoma protein 1 (RB1) in these Rb cells reduced cancer hallmarks such as proliferation, invasion, and spheroid formation and increased their sensitivity to chemotherapy drugs. Induction of HK1 was accompanied by a metabolic shift of the cells to glycolysis and a reduction in mitochondrial mass. Cytoplasmic HK1 bound Liver Kinase B1 and phosphorylated AMP-activated kinase-α (AMPKα Thr172), thereby reducing mitochondria-dependent energy production. We validated these findings in tumor samples from Rb patients compared to age-matched healthy retinae. HK1 or RB1 expression in Rb-/- cells led to a reduction in their respiratory capacity and glycolytic proton flux. HK1 overexpression reduced tumor burden in an intraocular tumor xenograft model. AMPKα activation by AICAR also enhanced the tumoricidal effects of the chemotherapeutic drug topotecan in vivo. Therefore, enhancing HK1 or AMPKα activity can reprogram cancer metabolism and sensitize Rb tumors to lower doses of existing treatments, a potential therapeutic modality for Rb.


Assuntos
Neoplasias da Retina , Retinoblastoma , Criança , Animais , Humanos , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Proteínas Quinases Ativadas por AMP , Fenótipo , Modelos Animais de Doenças , Neoplasias da Retina/genética , Neoplasias da Retina/patologia
12.
Exp Eye Res ; 233: 109542, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37331647

RESUMO

Retinoblastoma (Rb) is a rare malignant disorder affecting the developing retina of children under the age of five. Chemotherapeutic agents used for treating Rb have been associated with defects of the retinal pigment epithelium (RPE), such as hyperplasia, gliosis, and mottling. Herein, we have developed two pluripotent stem cell (PSC)-RPE models to assess the cytotoxicity of known Rb chemotherapeutics such as Melphalan, Topotecan and TW-37. Our findings demonstrate that these drugs alter the RPE by decreasing the monolayer barrier's trans-epithelial resistance and affecting the cells' phagocytic activity. Transcriptional analyses demonstrate an altered expression of genes involved in melanin and retinol processing, tight junction and apical-basal polarity pathways in both models. When applied within the clinical range, none of the drug treatments caused significant cytotoxic effects, changes to the apical-basal polarity, tight junction network or cell cycle. Together, our results demonstrate that although the most commonly used Rb chemotherapeutic drugs do not cause cytotoxicity in RPE, their application in vitro leads to compromised phagocytosis and strength of the barrier function, in addition to changes in gene expression that could alter the visual cycle in vivo. Our data demonstrate that widely used Rb chemotherapeutic drugs can have a deleterious impact on RPE cells and thus great care has to be exercised with regard to their delivery so the adjacent healthy RPE is not damaged during the course of tumor eradication.


Assuntos
Neoplasias da Retina , Retinoblastoma , Criança , Humanos , Epitélio Pigmentado da Retina/metabolismo , Retinoblastoma/tratamento farmacológico , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retina , Neoplasias da Retina/tratamento farmacológico , Neoplasias da Retina/genética , Neoplasias da Retina/metabolismo , Expressão Gênica , Diferenciação Celular
13.
Adv Biol Regul ; 88: 100964, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37004354

RESUMO

Small cell lung cancer (SCLC) often exhibits Rb deficiency, TRß and p130 deletion, and SKP2 amplification, suggesting TRß inactivation and SKP2 activation. It is reported that SKP2 targeted therapy is effective in some cancers in vitro and in vivo, but it is not reported for the treatment of SCLC and retinoblastoma. SKP2 is the synthetic lethal gene in SCLC and retinoblastoma, so SKP2 can be used for targeted therapy in SCLC and retinoblastoma. RB1 knockout mice develop several kinds of tumors, but Rb1 and SKP2 double knockout mice are healthy, suggesting that SKP2 targeted therapy may have significant effects on Rb deficient cancers with less side effects, and if successful in SCLC and retinoblastoma in vitro and in animal model, such compounds may be promising for the clinical treatment of SCLC, retinoblastoma, and variety of Rb deficient cancers. Previously our studies showed that retinoblastomas exhibit retinal cone precursor properties and depend on cone-specific thyroid hormone receptor ß2 (TRß2) and SKP2 signaling. In this study, we sought to suppress SCLC and retinoblastoma cell growth by SKP2 inhibitors as a prelude to targeted therapy in vitro and in vivo. We knocked down TRß2 and SKP2 or over-expressed p27 in SCLC and retinoblastoma cell lines to investigate SKP2 and p27 signaling alterations. The SCLC cell lines H209 as well as retinoblastoma cell lines Y79, WERI, and RB177 were treated with SKP2 inhibitor C1 at different concentrations, following which Western blotting, Immunostaining, and cell cycle kinetics studies were performed to study SKP2 and p27 expression ubiquitination, to determine impact on cell cycle regulation and growth inhibition. TRß2 knockdown in Y79, RB177 and H209 caused SKP2 downregulation and degradation, p27 up-regulation, and S phase arrest, whereas, SKP2 knockdown or p27 over-expression caused p27 accumulation and G1-S phase arrest. In the cell lines Y79, WERI, RB177, and H209 treatment with C1 caused SKP2 ubiquitination and degradation, p27 de-ubiquitination and accumulation, and cell growth arrest. SKP2 inhibitor C1 significantly suppressed retinoblastoma as well as SCLC cell growth by SKP2 degradation and p27 accumulation. In vivo study also showed inhibition of tumor growth with C1 treatment. Potential limitations of the success of such a therapeutic approach and its translational application in human primary tumors, and alternative approaches to overcome such limitations are briefly discussed for the treatment of retinoblastoma, SCLC and other RB-related cancers.


Assuntos
Neoplasias da Retina , Retinoblastoma , Camundongos , Animais , Humanos , Retinoblastoma/tratamento farmacológico , Retinoblastoma/genética , Retinoblastoma/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Linhagem Celular Tumoral , Ciclo Celular , Camundongos Knockout , Pulmão/patologia
14.
Sci China Life Sci ; 66(9): 2138-2151, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36949231

RESUMO

Retinoblastoma, the most prevalent primary intraocular tumor in children, leads to vision impairment, disability and even death. In addition to RB1 inactivation, MYCN activation has been documented as another common oncogenic alteration in retinoblastoma and represents one of the high-risk molecular subtypes of retinoblastoma. However, how MYCN contributes to the progression of retinoblastoma is still incompletely understood. Here, we report that MYCN upregulates YTHDF1, which encodes one of the reader proteins for N6-methyladenosine (m6A) RNA modification, in retinoblastoma. We further found that this MYCN-upregulated m6A reader functions to promote retinoblastoma cell proliferation and tumor growth in an m6A binding-dependent manner. Mechanistically, YTHDF1 promotes the expression of multiple oncogenes by binding to their mRNAs and enhancing mRNA stability and translation in retinoblastoma cells. Taken together, our findings reveal a novel MYCN-YTHDF1 regulatory cascade in controlling retinoblastoma cell proliferation and tumor growth, pinpointing an unprecedented mechanism for MYCN amplification and/or activation to promote retinoblastoma progression.


Assuntos
Neoplasias da Retina , Retinoblastoma , Criança , Humanos , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Proteína Proto-Oncogênica N-Myc/genética , Proteína Proto-Oncogênica N-Myc/metabolismo , Oncogenes , Neoplasias da Retina/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
15.
Biotechnol J ; 18(5): e2200518, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36808896

RESUMO

BACKGROUND: Retinoblastoma (Rb) is a rare cancer of the retina that occurs during early childhood. The disease is relatively rare but aggressive, accounting for ∼3% of childhood cancers. Treatment modalities encompass the administration of large doses of chemotherapeutic drugs, which result in multiple side-effects. Therefore, it is essential to have safe and effective newer therapies and suitable physiologically relevant, alternative-to-animal, in vitro cell culture-based models to enable rapid and efficient evaluation of potential therapies. METHODOLOGY: This investigation was focused on the development of a triple co-culture model comprising Rb, retinal epithelium, and choroid endothelial cells, using a protein coating cocktail, to recapitulate this ocular cancer under in vitro conditions. This resulting model was used for screening drug toxicity, based on the growth profile of Rb cells, using carboplatin as the model drug. Further, a combination of bevacizumab and carboplatin was evaluated using the developed model, to lower the concentration of carboplatin and thereby reduce its physiological side-effects. MAJOR RESULTS: The effect of drug treatment on the triple co-culture was assessed by increase in the apoptotic profile of Rb cells. Further, the barrier properties were found to be lower with a decrease in the angiogenetic signals that included expression of vimentin. Measurement of cytokine levels signified reduced inflammatory signals due to the combinatorial drug treatment. CONCLUSIONS: These findings validated that the triple co-culture Rb model was suitable for evaluating anti-Rb therapeutics and could thereby decrease the immense load on animal trials, which are the primary screens employed for evaluating retinal therapies.


Assuntos
Neoplasias da Retina , Retinoblastoma , Animais , Humanos , Retinoblastoma/tratamento farmacológico , Retinoblastoma/metabolismo , Carboplatina/uso terapêutico , Células Endoteliais/metabolismo , Retina/metabolismo , Neoplasias da Retina/tratamento farmacológico , Neoplasias da Retina/metabolismo
16.
Ocul Immunol Inflamm ; 31(3): 506-514, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35404750

RESUMO

OBJECTIVE: LINC00488 confers oncogenic activity in the progression of some tumors. Hence, the target of the study was about to specify LINC00488-mediated network in retinoblastoma (RB). METHODS: LINC00488 expression was tested in RB clinical tissues. siRNA targeting LINC00488 or miR-30a-5p mimic was introduced into RB cell line (Y79) to observe cellular biological functions. The relationship between LINC00488, miR-30a-5p and EPHB2 was verified. Afterward, the role of miR-30a-5p involved in RB through targeted regulation of EPHB2 was probed in vitro and in vivo. RESULTS: LINC00488 was induced in RB tissue and cells. LINC00488 knockdown or miR-30a-5p upregulation depressed the malignant activities of Y79 cells. LINC00488 could sponge miR-30a-5p that targeted EPHB2. EPHB2, and EPHB2 overexpression counteracted miR-30a-5p restoration-induced inhibition of Y79 cell development in vitro and in vivo. CONCLUSION: LINC00488 induces tumorigenicity in RB by binding to miR-30a-5p to target EPHB2, which may offer a new clue of RB treatment from an lncRNA-miRNA-mRNA network.


Assuntos
MicroRNAs , RNA Longo não Codificante , Receptor EphB2 , Neoplasias da Retina , Retinoblastoma , Humanos , Linhagem Celular Tumoral , Proliferação de Células , MicroRNAs/metabolismo , Neoplasias da Retina/metabolismo , Neoplasias da Retina/patologia , Retinoblastoma/metabolismo , Retinoblastoma/patologia , RNA Longo não Codificante/metabolismo , Receptor EphB2/metabolismo
17.
Klin Monbl Augenheilkd ; 240(7): 878-886, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34571550

RESUMO

BACKGROUND: The role of CD133 und ABCB5 is discussed in treatment resistance in several types of cancer. The objective of this study was to evaluate whether CD133+/ABCB5+ colocalization differs in untreated, in beam radiation treated, and in chemotherapy treated retinoblastoma specimens. Additionally, CD133, ABCB5, sphingosine kinase 1, and sphingosine kinase 2 gene expression was analyzed in WERI-RB1 (WERI RB1) and etoposide-resistant WERI RB1 subclones (WERI ETOR). METHODS: Active human untreated retinoblastoma specimens (n = 12), active human retinoblastoma specimens pretreated with beam radiation before enucleation (n = 8), and active human retinoblastoma specimens pretreated with chemotherapy before enucleation (n = 7) were investigated for localization and expression of CD133 and ABCB5 by immunohistochemistry. Only specimens with IIRC D, but not E, were included in this study. Furthermore, WERI RB1 and WERI ETOR cell lines were analyzed for CD133, ABCB5, sphingosine kinase 1, and sphingosine kinase 2 by the real-time polymerase chain reaction (RT-PCR). RESULTS: Immunohistochemical analysis revealed the same amount of CD133+/ABCB5+ colocalization islets in untreated and treated human retinoblastoma specimens. Quantitative RT-PCR analysis showed a statistically significant upregulation of CD133 in WERI ETOR (p = 0.002). No ABCB5 expression was detected in WERI RB1 and WERI ETOR. On the other hand, SPHK1 (p = 0.0027) and SPHK2 (p = 0.017) showed significant downregulation in WERI ETOR compared to WERI RB1. CONCLUSIONS: CD133+/ABCB5+ co-localization islets were noted in untreated and treated human retinoblastoma specimens. Therefore, we assume that CD133+/ABCB5+ islets might play a role in retinoblastoma genesis, but not in retinoblastoma treatment resistance.


Assuntos
Neoplasias da Retina , Retinoblastoma , Humanos , Retinoblastoma/genética , Retinoblastoma/tratamento farmacológico , Retinoblastoma/metabolismo , Etoposídeo/uso terapêutico , Neoplasias da Retina/genética
18.
Chem Biol Drug Des ; 101(3): 605-613, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36178076

RESUMO

This study aims to investigate cordycepin's effects on the proliferation, migration, and invasion of retinoblastoma (RB) cells and its regulatory mechanism. In this study, it was found that cordycepin inhibited RB cell proliferation, migration, and invasion in vitro, and pulmonary metastasis in vivo. c-Myc was a downstream target of cordycepin, and cordycepin significantly suppressed c-Myc expression, and c-Myc overexpression markedly counteracted the impacts of cordycepin on RB cell proliferation, migration, and invasion. c-Myc was positively correlated with the cell cycle pathway. Cordycepin restrained cyclin D1 expression, and c-Myc overexpression rescued this effect. In conclusion, cordycepin targets the c-Myc/cyclin D1 pathway, thereby suppressing the malignant biological behaviors of RB cells.


Assuntos
Neoplasias Pulmonares , Neoplasias da Retina , Retinoblastoma , Humanos , Ciclina D1 , Retinoblastoma/tratamento farmacológico , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Proliferação de Células , Neoplasias Pulmonares/tratamento farmacológico , Linhagem Celular Tumoral , Movimento Celular
19.
Exp Eye Res ; 226: 109340, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36476400

RESUMO

Retinoblastoma (RB) is the most common neoplasm found in the eye of children. There are increasing interests to develop targeted gene therapy for this disease. This study was performed to investigate the impact of long non-coding RNA (lncRNA) MEG3 on the biological features of RB cells. Vector overexpressing MEG3 was constructed and introduced into two RB cell lines. Transfected RB cells were assessed for proliferation, apoptosis, migration ability, expression levels of important genes in the PI3K/Akt/mTOR signaling pathway using qRT-PCR and Western blot analysis. Xenograft mouse models were constructed to determine the tumorigenicity of RB cells overexpressing MEG3. MEG3 mRNA level was significantly lower in RB cells than in non-cancer cells (p < 0.01). Overexpressing MEG3 resulted in significant reduction in cell proliferation (p < 0.05), migration (p < 0.01) and significant increase in apoptosis (p < 0.01). After overexpressing MEG3, p-PI3K, p-Akt and p-mTOR levels were significantly downregulated (p < 0.01). Furthermore, in the xenograft model, RB cells overexpressing MEG3 generated significantly smaller tumors as compared to RB cells that did not overexpress MEG3 (p < 0.05). Our data suggest that MEG3 increases apoptosis and reduces tumorigenicity of RB cells through inactivating the PI3K/Akt/mTOR pathway. Therefore, MEG3 could be further investigated as a potential new therapeutic agent and target for RB therapy.


Assuntos
RNA Longo não Codificante , Retinoblastoma , Animais , Humanos , Camundongos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias da Retina/genética , Neoplasias da Retina/metabolismo , Neoplasias da Retina/patologia , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo
20.
Funct Integr Genomics ; 23(1): 13, 2022 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-36547723

RESUMO

Retinoblastoma (RB), the most common malignant retinal tumor among children under 3 years old, is lethal if left untreated. Early diagnosis, together with timely and effective treatment, is important to improve retinoblastoma-related outcomes. Circular RNAs (circRNAs), a new class of non-coding RNAs with the capacity to regulate cellular activities, have great potential in retinoblastoma diagnosis and treatment. Recent studies have identified circular RNAs that regulate multiple cellular processes involved in retinoblastoma, including cell viability, proliferation, apoptosis, autophagy, migration, and invasion. Six circular RNAs (circ-FAM158A, circ-DHDDS, circ-E2F3, circ-TRHDE, circ-E2F5, and circ-RNF20) promote disease progression and metastasis in retinoblastoma and function as oncogenic factors. Other circular RNAs, such as circ-TET1, circ-SHPRH, circ-MKLN1, and circ-CUL2, play tumor suppressive roles in retinoblastoma. At present, the studies on the regulatory mechanism of circular RNAs in retinoblastoma are not very clear. The purpose of this review is to summarize recent studies on the functional roles and molecular mechanisms of circular RNAs in retinoblastoma and highlight novel strategies for retinoblastoma diagnosis, prognosis, and treatment.


Assuntos
MicroRNAs , RNA Circular , Neoplasias da Retina , Retinoblastoma , Criança , Pré-Escolar , Humanos , Apoptose , Linhagem Celular Tumoral , Proliferação de Células/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias da Retina/diagnóstico , Neoplasias da Retina/metabolismo , Neoplasias da Retina/terapia , Retinoblastoma/diagnóstico , Retinoblastoma/metabolismo , Retinoblastoma/terapia , RNA Circular/genética , RNA Circular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA