Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Hum Genet ; 143(2): 107-124, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38276976

RESUMEN

Ovarian cancer (OC) is a fatal gynecological disease that is often diagnosed at later stages due to its asymptomatic nature and the absence of efficient early-stage biomarkers. Previous studies have identified genes with abnormal expression in OC that couldn't be explained by methylation or mutation, indicating alternative mechanisms of gene regulation. Recent advances in human transcriptome studies have led to research on non-coding RNAs (ncRNAs) as regulators of cancer gene expression. Long non-coding RNAs (lncRNAs), a class of ncRNAs with a length greater than 200 nucleotides, have been identified as crucial regulators of physiological processes and human diseases, including cancer. Dysregulated lncRNA expression has also been found to play a crucial role in ovarian carcinogenesis, indicating their potential as novel and non-invasive biomarkers for improving OC management. However, despite the discovery of several thousand lncRNAs, only one has been approved for clinical use as a biomarker in cancer, highlighting the importance of further research in this field. In addition to their potential as biomarkers, lncRNAs have been implicated in modulating chemoresistance, a major problem in OC. Several studies have identified altered lncRNA expression upon drug treatment, further emphasizing their potential to modulate chemoresistance. In this review, we highlight the characteristics of lncRNAs, their function, and their potential to serve as tumor markers in OC. We also discuss a few databases providing detailed information on lncRNAs in various cancer types. Despite the promising potential of lncRNAs, further research is necessary to fully understand their role in cancer and develop effective strategies to combat this devastating disease.


Asunto(s)
Neoplasias Ováricas , ARN Largo no Codificante , Femenino , Humanos , ARN Largo no Codificante/genética , Resistencia a Antineoplásicos/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , ARN no Traducido , Biomarcadores de Tumor/genética
2.
Molecules ; 26(12)2021 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-34199287

RESUMEN

High-grade epithelial ovarian cancer is a fatal disease in women frequently associated with drug resistance and poor outcomes. We previously demonstrated that a marine-derived compound MalforminA1 (MA1) was cytotoxic for the breast cancer cell line MCF-7. In this study, we aimed to examine the effect of MA1 on human ovarian cancer cells. The potential cytotoxicity of MA1was tested on cisplatin-sensitive (A2780S) and cisplatin-resistant (A2780CP) ovarian cancer cell lines using AlamarBlue assay, Hoechst dye, flow cytometry, Western blot, and RT-qPCR. MA1 had higher cytotoxic activity on A2780S (IC50 = 0.23 µM) and A2780CP (IC50 = 0.34 µM) cell lines when compared to cisplatin (IC50 = 31.4 µM and 76.9 µM, respectively). Flow cytometry analysis confirmed the cytotoxic effect of MA1. The synergistic effect of the two drugs was obvious, since only 13% of A2780S and 7% of A2780CP cells remained alive after 24 h of treatment with both MA1 and cisplatin. Moreover, we examined the expression of bcl2, p53, caspase3/9 genes at RNA and protein levels using RT-qPCR and Western blot, respectively, to figure out the cell death mechanism induced by MA1. A significant down-regulation in bcl2 and p53 genes was observed in treated cells compared to non-treated cells (p < 0.05), suggesting that MA1 may not follow the canonical pathway to induce apoptosis in ovarian cancer cell lines. MalforminA1 showed promising anticancer activity by inducing cytotoxicity in cisplatin-sensitive and cisplatin-resistant cancer cell lines. Interestingly, a synergistic effect was observed when MA1 was combined with cisplatin, leading to it overcoming its resistance to cisplatin.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cisplatino/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Péptidos Cíclicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Femenino , Humanos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Péptidos Cíclicos/administración & dosificación
3.
BMC Cancer ; 20(1): 374, 2020 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366234

RESUMEN

BACKGROUND: The adhesion molecule, FAT4, has a tumor suppressor function with a critical role in the epithelial-to-mesenchymal-transition (EMT) and anti-malignant growth in several cancers. No study has investigated yet its role in epithelial ovarian cancer (EOC) progression. In the present study, we examined the role of FAT4 in proliferation and metastasis, and its mechanisms of interaction in these processes. METHODS: We have performed cell viability, colony formation, and invasion assays in ovarian cancer cells treated with siRNA to knockdown FAT4 gene expression. The regulatory effects of FAT4 on proteins involved in apoptotic, Wnt, Hippo, and retinoblastoma signaling pathways were evaluated by Western blotting following FAT4 repression. Also, 426 ovarian tumor samples and 88 non-tumor samples from the Gene Expression Profiling Interactive Analysis (GEPIA) database were analyzed for the expression of FAT4. Pearson's correlation was performed to determine the correlation between FAT4 and the E2F5, cyclin D1, cdk4, and caspase 9 expressions. RESULTS: Lower expression of FAT4 was observed in ovarian cancer cell lines and human samples as compared to non-malignant tissues. This down-regulation seems to enhance cell viability, invasion, and colony formation. Silencing FAT4 resulted in the upregulation of E2F5, vimentin, YAP, ß-catenin, cyclin D1, cdk4, and Bcl2, and in the downregulation of GSK-3-ß, and caspase 9 when compared to control. Furthermore, regulatory effects of FAT4 on the EMT and aggressive phenotype seem to occur through Hippo, Wnt, and cell cycle pathways. CONCLUSION: FAT4 downregulation promotes increased growth and invasion through the activation of Hippo and Wnt-ß-catenin pathways.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cadherinas/antagonistas & inhibidores , Factor de Transcripción E2F5/metabolismo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/patología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , beta Catenina/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Genes Supresores de Tumor , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Señalizadoras YAP
4.
Future Sci OA ; 9(1): FSO833, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37006229

RESUMEN

More than two-thirds of epithelial ovarian cancer (EOC) patients are diagnosed at advanced stages due to the lack of sensitive biomarkers. Currently, exosomes are intensively investigated as non-invasive cancer diagnostic markers. Exosomes are nanovesicles released in the extracellular milieu with the potential to modulate recipient cells' behavior. EOC cells release many altered exosomal cargoes that exhibit clinical relevance to tumor progression. Exosomes represent powerful therapeutic tools (drug carriers or vaccines), posing a promising option in clinical practice for curing EOC in the near future. In this review, we highlight the importance of exosomes in cell-cell communication, epithelial-mesenchymal transition (EMT), and their potential to serve as diagnostic and prognostic factors, particularly in EOC.


Exosomes are nanovesicles released in the extracellular milieu by diverse cell types and serve as carriers of proteins, and nucleic acids, with the potential to modulate the recipient cells. Understanding the functions of exosomes as messengers between cancer and healthy cells unfolds new avenues in intercellular signaling mechanisms. In this review, we have highlighted the importance of exosomes in cell­cell communication and their potential to serve as diagnostic and prognostic factors in EOC. Furthermore, we believe that exosomes could represent a powerful therapeutic tool due to their ability to function as drug carriers or vaccines posing a promising option in curing EOC.

5.
Asian Pac J Cancer Prev ; 24(5): 1583-1590, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37247277

RESUMEN

BACKGROUND: Chromatin immunoprecipitation (ChIP) analysis revealed that the FBXW7 gene and the long non-coding RNA (LINC01588) are potential candidates in epithelial ovarian cancer (EOC) pathogenesis. However, their exact role in EOC is not yet known. Thus, the present study sheds light on the impact of the mutations/ methylation status of the FBXW7 gene. MATERIALS AND METHODS: We used public databases to assess the correlation between mutations/ methylation status and the FBXW7 expression. Furthermore, we performed Pearson's correlation analysis between the FBXW7 gene and LINC01588. We performed gene panel exome sequencing and Methylation-specific PCR (MSP) in HOSE 6-3, MCAS, OVSAHO, and eight EOC patients' samples to validate the bioinformatics results. RESULTS: The FBXW7 gene was less expressed in EOC, particularly in stages III and IV, compared to healthy tissues. Furthermore, bioinformatics analysis, gene panel exome sequencing, and MSP revealed that the FBXW7 gene is neither mutated nor methylated in EOC cell lines and tissues, suggesting alternative mechanisms for FBXW7 gene regulation. Interestingly, Pearson's correlation analysis showed an inverse, significant correlation between the FBXW7 gene and LINC01588  expression, suggesting a potential regulatory role of LINC01588. CONCLUSION: Neither mutations nor methylation is the causative mechanism for the FBXW7 downregulation in EOC, suggesting alternative means involving the lncRNA LINC01588.


Asunto(s)
Neoplasias Ováricas , ARN Largo no Codificante , Humanos , Femenino , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Metilación de ADN , Regiones Promotoras Genéticas , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Carcinoma Epitelial de Ovario/patología , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , ARN Largo no Codificante/genética
6.
Hum Cell ; 36(2): 762-774, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36513868

RESUMEN

Epithelial Ovarian Cancer (EOC) is a heterogeneous disease usually diagnosed at advanced stages. Therefore, early detection is crucial for better survival. Despite the advances in ovarian research, mechanisms underlying EOC carcinogenesis are not elucidated. We performed chromatin immunoprecipitation sequencing to identify genes regulated by E2F5, a transcription factor involved in ovarian carcinogenesis. Results revealed several putative candidate genes (115 protein-coding genes, 20 lncRNAs, 6 pseudogenes, and 4 miRNAs). A literature review and bioinformatics analysis of these genes revealed a novel lncRNA candidate (LINC01465) in EOC. We validated LINC01465 by quantifying its expression in EOC cell lines and selected OVSAHO and SKOV3 as a model with high LINC01465 levels. We silenced LINC01465 and performed proliferation, wound healing, invasion, and drug resistance assays. Knocking-down LINC01465 resulted in reduced migration, suggesting potential involvement in EOC. Furthermore, to identify the significance of LINC01465 in chemoresistance, we assessed the LINC01465 levels in A2780 S cells treated with malformin, which revealed higher LINC01465 expression as compared to untreated A2780S cells implying the involvement of LINC01465 in cell death. Thus, this study unraveled the repertoire of E2F5 regulated candidate genes and suggested a putative role of LINC01465 in malformin-induced cell death in EOC.


Asunto(s)
Neoplasias Ováricas , ARN Largo no Codificante , Femenino , Humanos , Carcinogénesis , Carcinoma Epitelial de Ovario/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , ARN Largo no Codificante/genética
7.
Hum Cell ; 35(4): 1114-1125, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35596898

RESUMEN

Gastric cancer (GC) is ranked the third leading cause of cancer-related deaths worldwide. Mutations and epigenetic alterations in several essential genes, including p53, KRAS, PIK3CA, FAT4 and ARID1A, are often reported. Furthermore, loss of SOCS3 expression was reported in GC, suggesting its tumor suppressor role. To assess the mutational and methylation status of SOCS3, we performed gene panel exome sequencing on 47 human GC samples. The SOCS3 gene was rarely mutated, suggesting alternative regulation mechanisms, such as promoter hypermethylation and/or long non-coding RNAs (lncRNAs). We first explored SOCS3 promoter methylation status in 44 human GC samples by methylation-specific PCR (MS-PCR). Thirteen out of forty-four patients (29.5%) displayed a methylation pattern. Then, to see whether SOCS3 expression is silenced by CpG methylation, we examined publicly available databases (cbioportal and The Cancer Genome Atlas (TCGA)). The analysis revealed ß values lower than 0.1, indicating hypo-methylation in healthy and GC samples. Moreover, moderate methylation (ß < 0.4) and high methylation (ß > 0.4) did not affect the free survival, suggesting that methylation is unlikely to be the mechanism ruling SOCS3 silencing in GC. Next, to assess the regulatory effects of lncRNAs on SOCS3, we silenced the AC125807.2-lncRNA and quantified the SOCS3 gene expression in AGS and NCI-N87 gastric cancer cell line. SOCS3 was found to be downregulated following AC125807.2-lncRNA silencing in AGS cells, suggesting the potential implication of lncRNA AC125807.2 in SOCS3 regulation. However, in NCI-N87 cells, there was no significant change in SOCS3 expression. In conclusion, neither mutations nor hypermethylation was associated with the SOCS3 downregulation in GC, and alternative mechanisms, including non-coding RNAs-mediated gene silencing, may be proposed.


Asunto(s)
ARN Largo no Codificante , Neoplasias Gástricas , Línea Celular Tumoral , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen , Humanos , Mutación/genética , ARN Largo no Codificante/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína 3 Supresora de la Señalización de Citocinas/genética
8.
Asian Pac J Cancer Prev ; 23(8): 2661-2669, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-36037120

RESUMEN

OBJECTIVE: Ovarian cancer is one of the leading causes of cancer-related mortality in women, and is often associated with drug resistance. Therefore, finding effective drugs, including naturally derived compounds, is urgently needed. Herein, we aimed to test the anti-cancer potential of gallic acid monohydrate (GA) and its congeners on cisplatin-sensitive (A2780S), and resistant (A2780CP) ovarian cancer and normal ovarian (HOSE6-3) cell lines. METHODS: Cytotoxicity was assessed by AlamarBlue and CCK08 assays by exposing cells to different concentrations of cisplatin (0-21µg/mL), GA and its congeners (0-100µg/mL), and a combination of GA and cisplatin. Apoptosis was estimated by Hoechst stain and monitoring the relative RNA expression of the apoptotic effector caspase-3 using qRT-PCR. RESULTS: GA decreased cell viability in a concentration-dependent manner in all cell lines, with an IC50 of 19.39µg/mL (A2780S), 35.59 µg/mL (A2780CP), and 49.32µg/mL (HOSE6-3). GA displayed higher cytotoxicity than its congeners. An apoptotic rate estimation of approximately 20% and 30% was obtained in A2780S and A2780CP. While the cytotoxicity observed with cisplatin and GA was comparable, combining the two enhanced the cytotoxicity significantly, especially in the A2780CP cell line (p<0.05). CONCLUSION: These data suggest that GA may help overcome the resistance. Hence, the cytotoxic effects of GA, especially on chemo-resistant ovarian cancer cells merit further investigation.
.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos , Femenino , Ácido Gálico/farmacología , Humanos , Neoplasias Ováricas/metabolismo
9.
Sultan Qaboos Univ Med J ; 21(4): 632-634, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34888085

RESUMEN

OBJECTIVES: New compounds are needed to overcome the resistance to commonly used cytotoxic chemotherapy for epithelial ovarian cancer. Marine sponges are a rich source of diverse chemical compounds and hymenialdisine has been found to have antiproliferative effects. This study aimed to investigate the cytotoxic effect of hymenialdisine in cisplatin-sensitive and cisplatin resistant ovarian cancer cell lines. METHODS: This study took place at Sultan Qaboos University, Muscat, Oman between August and November, 2019. The anti-cancer effects of hymenialdisine or cisplatin were assessed using treating cells with different concentrations of hymenialdisine and cisplatin. Cell viability was determined using the AlamarBlue® Assay. RESULTS: The half-maximal inhibitory concentration (IC50) of cisplatin was estimated at 31.4 µM for A2780S and 76.9 µM for A2780CP, whereas the IC50 of hymenialdisine was evaluated at 146.8 µM for A2780S cells. Despite the higher concentrations of hymenialdisine (up to 300 µM), IC50 could not be determined for the A2780CP cell line. CONCLUSION: When compared to cisplatin, hymenialdisine was less toxic against both A2780S and A2780CP ovarian cancer cell lines.


Asunto(s)
Cisplatino , Neoplasias Ováricas , Azepinas , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Pirroles
10.
Genet Test Mol Biomarkers ; 25(3): 179-186, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33734894

RESUMEN

Background: E2F5 is a transcription factor that is overexpressed in the early stages of ovarian cancer and has been suggested as a potential biomarker for early detection. In this study, we aimed to examine the role of E2F5 in invasion and proliferation of ovarian cancer cells. Materials and Methods: We performed cell viability, colony formation, and invasion assays using ovarian cancer cells treated with siRNA to knock down the E2F5 gene. The regulatory effects of E2F5 on proteins involved in the apoptotic, Wnt, Hippo, and retinoblastoma signaling pathways were evaluated by western blotting following E2F5 repression. In addition, we analyzed data available on Gene Expression Profiling Interactive Analysis for correlations between E2F5 and YAP, ß-catenin, cyclin D1, cdk4, and caspase-9. Results: E2F5 was highly expressed in ovarian cancer cell lines and samples when compared to the nonmalignant tissues. Downregulation of E2F5 inhibited cell viability and invasion and promoted the phosphorylation of YAP, GSK-3-ß, ß-catenin, and retinoblastoma. However, cyclin D1, cdk4, and caspase-9 were downregulated when compared to control. Conclusion: Overall, E2F5 promotes ovarian carcinogenesis via the regulation of Hippo and Wnt pathways.


Asunto(s)
Factor de Transcripción E2F5/metabolismo , Neoplasias Ováricas/metabolismo , Apoptosis/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Ciclina D1/genética , Bases de Datos Genéticas , Factor de Transcripción E2F5/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Vía de Señalización Hippo , Humanos , Invasividad Neoplásica/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
11.
Endocr Connect ; 7(1): 65-77, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29138235

RESUMEN

In Oman, breast cancer is most common, representing approximately more than 25% of all cancers in women. Relatively younger populations of patients (25-40 years) present surprisingly with an aggressive phenotype and advanced tumor stages. In this study, we investigated differential gene expressions in Luminal A, Luminal B, triple-negative and Her2+ breast cancer subtypes and compared data to benign tumor samples. We identified a potential candidate gene BRIP1, showing differential expression in the four breast cancer subtypes examined, suggesting that BRIP1 has the profile of a useful diagnostic marker, suitable for targeted therapeutic intervention. RT-qPCR and Western blotting analysis showed higher BRIP1 expression in luminal samples as compared to triple-negative subtype patient's samples. We further screened BRIP1 for eventual mutations/SNPs/deletions by sequencing the entire coding region. Four previously identified polymorphisms were detected, one within the 5'-UTR region (c.141-64G > A) and three in the BRCA-binding domain (c.2755T > C, c.2647G > A and c.3411T > C). Kaplan-Meier analysis revealed that patients with overexpression of BRIP1 displayed a poor survival rate (P < 0.05). BRIP1 has a dual function of an oncogene and a tumor suppressor gene in addition to its role as a potential biomarker to predict survival and prognosis. Data obtained in this study suggest that BRIP1 can plausibly have an oncogenic role in sporadic cancers.

12.
Mol Clin Oncol ; 7(1): 3-14, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28685067

RESUMEN

Breast and ovarian cancer are heterogeneous diseases. While breast cancer accounts for 25% of cancers worldwide, ovarian cancer accounts for 3.5% of all cancers and it is considered to be the most lethal type of cancer among women. In Oman, breast cancer accounts for 25% and ovarian cancer for 4.5% of all cancer cases. Various risk factors, including variable biological and clinical traits, are involved in the onset of breast and ovarian cancer. Although highly developed diagnostic and therapeutic methods have paved the way for better management, targeted therapy against specific biomarkers has not yet shown any significant improvement, particularly in triple-negative breast cancer and epithelial ovarian cancer, which are associated with high mortality rates. Thus, elucidating the mechanisms underlying the pathology of these diseases is expected to improve their prevention, prognosis and management. The aim of the present study was to provide a comprehensive review and updated information on genomics and proteomics alterations associated with cancer pathogenesis, as reported by several research groups worldwide. Furthermore, molecular research in our laboratory, aimed at identifying new pathways involved in the pathogenesis of breast and ovarian cancer using microarray and chromatin immunoprecipitation (ChIP), is discussed. Relevant candidate genes were found to be either up- or downregulated in a cohort of breast cancer cases. Similarly, ChIP analysis revealed that relevant candidate genes were regulated by the E2F5 transcription factor in ovarian cancer tissue. An ongoing study aims to validate these genes with a putative role as biological markers that may contribute to the development of targeted therapies for breast and ovarian cancer.

13.
Mutat Res ; 601(1-2): 46-50, 2006 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16814811

RESUMEN

To determine whether PAX-2 and PAX-8 are involved in Wilms tumor (WT) pathogenesis, we sought mutations in these two genes in 99 Wilms tumors of favorable histology. We screened the entire protein coding sequences as well as the intronic regions adjacent to exons, using denaturing HPLC followed by sequencing of samples displaying abnormal chromatograms. In PAX-2, a silent polymorphism was found within exon 2 and exon 8 in 1% and 21% of cases, respectively. Three apparently silent polymorphisms were also found in PAX-8, two in exon 5 (2 of 99 cases or 2%) and one in exon 6 (22 of 99 cases or 22%), all of which were located 3' to the exons. In conclusion, no evidence for disease causing mutation was found using this technique, and so the direct involvement of either of these two genes in WT is unlikely.


Asunto(s)
Neoplasias Renales/genética , Factor de Transcripción PAX2/genética , Factores de Transcripción Paired Box/genética , Tumor de Wilms/genética , Cromatografía Líquida de Alta Presión/métodos , Exones/genética , Frecuencia de los Genes , Humanos , Mutación/genética , Factor de Transcripción PAX8 , Polimorfismo Genético/genética , Análisis de Secuencia de ADN/métodos
14.
Sultan Qaboos Univ Med J ; 16(2): e168-74, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27226907

RESUMEN

OBJECTIVES: Marine organisms are a rich source of bioactive molecules with potential applications in medicine, biotechnology and industry; however, few bioactive compounds have been isolated from organisms inhabiting the Arabian Gulf and the Gulf of Oman. This study aimed to isolate and screen the anti-cancer activity of compounds and extracts from 40 natural products of marine organisms collected from the Gulf of Oman. METHODS: This study was carried out between January 2012 and December 2014 at the Sultan Qaboos University, Muscat, Oman. Fungi, bacteria, sponges, algae, soft corals, tunicates, bryozoans, mangrove tree samples and sea cucumbers were collected from seawater at Marina Bandar Al-Rowdha and Bandar Al-Khayran in Oman. Bacteria and fungi were isolated using a marine broth and organisms were extracted with methanol and ethyl acetate. Compounds were identified from spectroscopic data. The anti-cancer activity of the compounds and extracts was tested in a Michigan Cancer Foundation (MCF)-7 cell line breast adenocarcinoma model. RESULTS: Eight pure compounds and 32 extracts were investigated. Of these, 22.5% showed strong or medium anti-cancer activity, with malformin A, kuanoniamine D, hymenialdisine and gallic acid showing the greatest activity, as well as the soft coral Sarcophyton sp. extract. Treatment of MCF-7 cells at different concentrations of Sarcophyton sp. extracts indicated the induction of concentration-dependent cell death. Ultrastructural analysis highlighted the presence of nuclear fragmentation, membrane protrusion, blebbing and chromatic segregation at the nuclear membrane, which are typical characteristics of cell death by apoptosis induction. CONCLUSION: Some Omani marine organisms showed high anti-cancer potential. The efficacy, specificity and molecular mechanisms of anti-cancer compounds from Omani marine organisms on various cancer models should be investigated in future in vitro and in vivo studies.

15.
Saudi Med J ; 37(10): 1068-75, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27652356

RESUMEN

OBJECTIVES: To identify genetic defects in an Omani family diagnosed with deafness.  METHODS: A cross-sectional association study was conducted at the Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoud, Oman and the Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium between August 2010 and September 2014. Microsatellites markers for nine non-syndromic genes were used to genotype the defective locus using the extracted DNA from family members. Sanger sequencing method was used to identify the disease causative mutation. Eazy linkage 5.05 was used to calculate the logarithm of odds score. Lasergene suite was used to detect the mutation position, and Phyre2, SMART, Rasmol, and GOR IV were used to predict the effects of the defect on protein structure and function.  RESULTS: The disease was linked to markers located on chromosome-2 and covering the OTOF (DFNB9) gene. A novel missense mutation that changed nucleotide C to G at position c.1469 and consequently the amino acid Proline to Arginine (P490R) on exon 15 was detected. Protein modeling analysis revealed the impact of the mutation on protein structure and the relevant C2C domain. The mutation seems to create a new protein isoform homologous to the complement component C1q.  CONCLUSION: These findings suggest that the mutation found in C2C domain of the OTOF gene is likely to cause deafness in the studied family reflecting the importance of C2 domains of otoferlin in hearing loss.


Asunto(s)
Pérdida Auditiva Central/complicaciones , Pérdida Auditiva/genética , Proteínas de la Membrana/genética , Mutación Missense , Estudios Transversales , Femenino , Pérdida Auditiva/complicaciones , Humanos , Masculino , Omán , Linaje
16.
Front Biosci (Elite Ed) ; 8(1): 40-5, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26709644

RESUMEN

HOXB13, a member of the homeobox proteins family, is a key regulator of the epithelial differentiation in the prostate gland. HOXB13 is overexpressed during malignant progression of the prostatic tissue and suspected to contribute in the pathogenesis of the prostate gland. In androgen deprived conditions, HOXB13 is thought to act through inhibition of the tumour suppressor protein p21. Since HOXB13 has a multifaceted role in ventral prostate development, its critical partners in the cascade need to be elucidated for a further understanding of its role in prostate malignancy. In this report, we review the functions attributed to HOXB13, by highlighting the most recent findings supporting the hypothesis that HOXB13 might serve as a novel biomarker for the prognosis of prostate cancer.


Asunto(s)
Biomarcadores de Tumor/fisiología , Proteínas de Homeodominio/fisiología , Neoplasias de la Próstata/patología , Diferenciación Celular , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Proteínas de Homeodominio/química , Humanos , Masculino , Neovascularización Patológica , Próstata/patología , Próstata/fisiopatología , Neoplasias de la Próstata/irrigación sanguínea , Piel/fisiopatología
17.
Front Biosci (Elite Ed) ; 7(2): 208-14, 2015 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-25553374

RESUMEN

The Pim-1 gene encodes for a proto-oncogenic serine/threonine protein kinase and is generally involved in cytokine signaling as well as in various signaling pathways regulating cell cycle and apoptosis. Pim-1 kinase plays a role in the development of various tumors mainly, prostate cancer, Burkitt's lymphoma, oral cancer and various other hematopoietic lymphomas. This review will focus on the importance and mechanisms of Pim-1 in prostate cancer and the potential clinical relevance of its various inhibitors.


Asunto(s)
Carcinoma/etiología , Neoplasias de la Próstata/etiología , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Animales , Carcinoma/enzimología , Humanos , Masculino , Estructura Molecular , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/genética
18.
Invest Ophthalmol Vis Sci ; 45(11): 3904-13, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15505035

RESUMEN

PURPOSE: To overcome the problem of antibody availability, often encountered during chromatin immunoprecipitation (ChIP) assays, nickel agarose-based chromatin enrichment (NACE) was developed. Based on the affinity of (His)-6-tagged proteins for the nickel ion, this modified form of ChIP allows the isolation of chromatin in the absence of specific antibodies. METHODS: Nonpigmented ciliary epithelium cells were transfected with (His)-6-tagged FOXC1. FOXC1-enriched chromatin complexes were isolated by using the tight electrostatic interaction between histidine residues of the recombinant FOXC1 protein and nickel. One hundred fifty NACE-enriched clones were sequenced and subjected to in silico and biochemical analyses. RESULTS: Twenty-six clones were detected near known genes: Eight were near predicted but uncharacterized genes, eight were within areas where neither known nor predicted genes have yet been mapped, four were chimeric, and the rest were either repetitive (n=81) or poor-quality (n=23) sequences. Twenty of the 26 known genes were expressed in the eye. Five of the NACE-enriched clones (BMP2K, DACH, FVT-1, SIX-1, and PGE-2 receptor), as well as nine clones selected from the literature, were validated by PCR amplification in two independent lots of NACE-enriched chromatin. All five NACE-selected genes were detected in two independent assays, as well as four (BMP7, SMAD2, TGF-B1, and WNT6) of the nine genes selected from the literature, consistent with these genes' being regulated by FOXC1. CONCLUSIONS: NACE is a useful technique allowing specific chromatin enrichment in cases where antibodies are unavailable. Specific recovery of PTGER, DACH1, WNT6, and FVT-1 implicates FOXC1 in a variety of cellular events including modulation of intraocular pressure, cell cycle, ocular development, and oncogenesis.


Asunto(s)
Cromatina/metabolismo , Cuerpo Ciliar/metabolismo , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Factores de Transcripción/fisiología , Cromatina/aislamiento & purificación , Cuerpo Ciliar/citología , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead , Marcación de Gen , Humanos , Níquel , Reacción en Cadena de la Polimerasa , Sefarosa , Transfección
19.
Virus Res ; 93(1): 1-12, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12727337

RESUMEN

Up to 50% of the transitional cell carcinomas (TCC) express an activated EGF pathway involving MAP/MEK and RAF kinase thus providing a novel means to selectively eliminate transformed cells expressing such proteins. This EGF pathway expression phenotype was also confirmed in our MGH-U3 and room temperature-112 human TCC cell lines, which makes them a suitable model target for the reovirus oncolysis. We report here on an in vitro assay of co-culture spheroids using either human or rat TCC cells with their corresponding fibroblasts to examine the potential of viral selective lysis for TCC. Reovirus, a respiratory enteric orphan virus, which mammals are exposed to early in life, was used in this study. Selective killing of transformed versus normal cells was assayed by time-lapse photography, vital dye staining, immunohistochemistry, and MTT assay. In this in vitro bladder cancer model, reovirus selectively destroyed the transformed cells by lysis or induction of apoptosis. Based on these findings we have initiated an in vivo pre-clinical study on intravesical administration of reovirus in an animal model to further explore the effect of reovirus-mediated oncolysis of TCC.


Asunto(s)
Carcinoma de Células Transicionales/patología , Carcinoma de Células Transicionales/virología , Reoviridae/patogenicidad , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/virología , Carcinoma de Células Transicionales/ultraestructura , Supervivencia Celular , Técnicas de Cocultivo , Fibroblastos/ultraestructura , Fibroblastos/virología , Humanos , Inmunohistoquímica , Infecciones por Reoviridae/patología , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/ultraestructura
20.
Urol Oncol ; 7(3): 125-32, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12474546

RESUMEN

Photodynamic therapy (PDT) may be an attractive option for treatment of early stage prostate cancer. Aminolevulinic acid (ALA) acts as a prodrug leading to a selective accumulation of a photosensitizer, protoporphyrin IX (PpIX), in epithelial cells. We investigated the efficacy of ALA-mediated PDT for rat R3327-H prostate cancer, compared with the AY-27 bladder tumor. Rats bearing either AY-27 or R3327-H tumors were randomized to different groups when their tumors reached approximately 1000 mm3. At the day of PDT, animals were administered 500 mg/kg ALA intravenously 4 hours prior to laser therapy. The argon-pumped dye laser light (630 nm) was coupled to multiple quartz fibers with cylindrical diffusing tips, which were inserted into the tumor in icosahedral pattern. Light exposure was varied to yield doses of 1000 to 3000 J/tumor. Animals bearing R3327-H tumors were imaged with 99mTc-HMPAO scintigraphy to evaluate tumor perfusion changes induced by PDT. There was a light-dose dependent tumor response in both tumor models. The mean time for R3327-H tumor to re-grow to 4 x treatment volume was 79.7 days in the control group (light only), 159 days in 1000 J group, and 169 days in 2000 J group (P < 0.05). Tumors treated with 3000 J were clinically cured (P < 0.01). Likewise, for AY-27 tumors, the average time to re-grow to 4 x treatment volume was 13.7 days in the control group, 179.3, 183.3, and 185.7 days in groups of 1000, 1500, and 2000 J (P < 0.05), respectively. Tumors treated with 3000 J were clinically cured (P < 0.01). 99mTc-HMPAO scintigraphy demonstrated a mild perfusion impairment following PDT. Interstitial PDT with ALA/PpIX is equally effective in treating prostate cancer and TCC in these heterotopic rat models.


Asunto(s)
Ácido Aminolevulínico/uso terapéutico , Fotoquimioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Adenocarcinoma/diagnóstico por imagen , Adenocarcinoma/tratamiento farmacológico , Animales , Masculino , Neoplasias de la Próstata/diagnóstico por imagen , Cintigrafía , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/diagnóstico por imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA