Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Pathol Res Pract ; 248: 154618, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37331185

RESUMEN

Globally, prostate cancer (PC) is leading cause of cancer-related mortality in men worldwide. Despite significant advances in the treatment and management of this disease, the cure rates for PC remains low, largely due to late detection. PC detection is mostly reliant on prostate-specific antigen (PSA) and digital rectal examination (DRE); however, due to the low positive predictive value of current diagnostics, there is an urgent need to identify new accurate biomarkers. Recent studies support the biological role of microRNAs (miRNAs) in the initiation and progression of PC, as well as their potential as novel biomarkers for patients' diagnosis, prognosis, and disease relapse. In the advanced stages, cancer-cell-derived small extracellular vesicles (SEVs) may constitute a significant part of circulating vesicles and cause detectable changes in the plasma vesicular miRNA profile. Recent computational model for the identification of miRNA biomarkers discussed. In addition, accumulating evidence indicates that miRNAs can be utilized to target PC cells. In this article, the current understanding of the role of microRNAs and exosomes in the pathogenesis and their significance in PC prognosis, early diagnosis, chemoresistance, and treatment are reviewed.


Asunto(s)
MicroARNs , Neoplasias de la Próstata , Masculino , Humanos , MicroARNs/genética , Biomarcadores de Tumor/genética , Detección Precoz del Cáncer , Recurrencia Local de Neoplasia , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Pronóstico
2.
Front Pharmacol ; 13: 823572, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35250573

RESUMEN

Gynecologic cancer is one of the main causes of death in women. In this type of cancer, several molecules (oncogenes or tumor suppressor genes) contribute to the tumorigenic process, invasion, metastasis, and resistance to treatment. Based on recent evidence, the detection of molecular changes in these genes could have clinical importance for the early detection and evaluation of tumor grade, as well as the selection of targeted treatment. Researchers have recently focused on cancer stem cells (CSCs) in the treatment of gynecologic cancer because of their ability to induce progression and recurrence of malignancy. This has highlighted the importance of a better understanding of the molecular basis of CSCs. The purpose of this review is to focus on the molecular mechanism of gynecologic cancer and the role of CSCs to discover more specific therapeutic approaches to gynecologic cancer treatment.

3.
J Oncol ; 2021: 3408937, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721577

RESUMEN

Ovarian cancer is the major cause of gynecologic cancer-related mortality. Regardless of outstanding advances, which have been made for improving the prognosis, diagnosis, and treatment of ovarian cancer, the majority of the patients will die of the disease. Late-stage diagnosis and the occurrence of recurrent cancer after treatment are the most important causes of the high mortality rate observed in ovarian cancer patients. Unraveling the molecular mechanisms involved in the pathogenesis of ovarian cancer may help find new biomarkers and therapeutic targets for ovarian cancer. MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression, mostly at the posttranscriptional stage, through binding to mRNA targets and inducing translational repression or degradation of target via the RNA-induced silencing complex. Over the last two decades, the role of miRNAs in the pathogenesis of various human cancers, including ovarian cancer, has been documented in multiple studies. Consequently, these small RNAs could be considered as reliable markers for prognosis and early diagnosis. Furthermore, given the function of miRNAs in various cellular pathways, including cell survival and differentiation, targeting miRNAs could be an interesting approach for the treatment of human cancers. Here, we review our current understanding of the most updated role of the important dysregulation of miRNAs and their roles in the progression and metastasis of ovarian cancer. Furthermore, we meticulously discuss the significance of miRNAs as prognostic and diagnostic markers. Lastly, we mention the opportunities and the efforts made for targeting ovarian cancer through inhibition and/or stimulation of the miRNAs.

4.
Sci Rep ; 11(1): 10524, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34006887

RESUMEN

Circulating tumor cells (CTCs) have recently been considered as new prognostic and diagnostic markers for various human cancers; however, their significance in epithelial ovarian cancer (EOC) remains to be elucidated. In this study, using quantitative real-time PCR, we evaluated the expression of EPCAM, MUC1, CEA, HE4 and CA125 mRNAs, as putative markers of CTCs, in the blood of 51 EOC patients before and/or after adjuvant chemotherapy. Our results demonstrated that, before chemotherapy, the expression of EPCAM, MUC1, CEA and HE4 mRNAs were correlated to each other. CEA expression was correlated with tumor stage (r = 0.594, p = 0.000) before chemotherapy, whereas its expression after chemotherapy was correlated with serum levels of CA125 antigen (r = 0.658, p = 0.000). HE4 mRNA showed the highest sensitivity both before and after chemotherapy (82.98% and 85.19%, respectively) and the persistence of this marker after chemotherapy was associated with advanced disease stage. The expression of CA125 mRNA had negative correlation with the other markers and with tumor stage and therapy response (evaluated by the measurement of serum CA125 antigen). Collectively, our results indicated a better clinical significance of tumor-specific markers (CEA and HE4 mRNAs) compared to epithelial-specific markers (EPCAM and MUC1 mRNAs).


Asunto(s)
Biomarcadores de Tumor/sangre , Carcinoma Epitelial de Ovario/sangre , Quimioterapia Adyuvante , Neoplasias Ováricas/sangre , Neoplasias Ováricas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Femenino , Humanos , Persona de Mediana Edad , Células Neoplásicas Circulantes/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Curr Pharm Des ; 27(28): 3161-3169, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33902409

RESUMEN

DNA damage usually happens in all cell types, which may originate from endogenous sources (i.e., DNA replication errors) or be emanated from radiations or chemicals. These damages range from changes in few nucleotides to significant structural abnormalities on chromosomes and, if not repaired, could disturb the cellular homeostasis or cause cell death. As the most significant response to DNA damage, DNA repair provides biological pathways by which DNA damages are corrected and returned into their natural circumstance. However, an aberration in the DNA repair mechanisms may result in genomic and chromosomal instability and the accumulation of mutations. The activation of oncogenes and/or inactivation of tumor suppressor genes is a serious consequence of genomic and chromosomal instability and may bring the cells into a cancerous phenotype. Therefore, genomic and chromosomal instability is usually considered a crucial factor in carcinogenesis and an important hallmark of various human malignancies. In the present study, we review our current understanding of the most updated mechanisms underlying genomic instability in cancer and discuss the potential promises of these mechanisms in finding new targets for the treatment of cancer.


Asunto(s)
Inestabilidad Genómica , Neoplasias , Inestabilidad Cromosómica/genética , Daño del ADN , Reparación del ADN/genética , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética
6.
Cell Oncol (Dordr) ; 43(4): 515-538, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32418122

RESUMEN

BACKGROUND: Ovarian cancer is the most lethal gynecologic cancer and the fifth leading cause of cancer-related mortality in women worldwide. Despite various attempts to improve the diagnosis and therapy of ovarian cancer patients, the survival rate for these patients is still dismal, mainly because most of them are diagnosed at a late stage. Up to 90% of ovarian cancers arise from neoplastic transformation of ovarian surface epithelial cells, and are usually referred to as epithelial ovarian cancer (EOC). Unlike most human cancers, which are disseminated through blood-borne metastatic routes, EOC has traditionally been thought to be disseminated through direct migration of ovarian tumor cells to the peritoneal cavity and omentum via peritoneal fluid. It has recently been shown, however, that EOC can also be disseminated through blood-borne metastatic routes, challenging previous thoughts about ovarian cancer metastasis. CONCLUSIONS: Here, we review our current understanding of the most updated cellular and molecular mechanisms underlying EOC metastasis and discuss in more detail two main metastatic routes of EOC, i.e., transcoelomic metastasis and hematogenous metastasis. The emerging concept of blood-borne EOC metastasis has led to exploration of the significance of circulating tumor cells (CTCs) as novel and non-invasive prognostic markers in this daunting cancer. We also evaluate the role of tumor stroma, including cancer associated fibroblasts (CAFs), tumor associated macrophages (TAMs), endothelial cells, adipocytes, dendritic cells and extracellular matrix (ECM) components in EOC growth and metastasis. Lastly, we discuss therapeutic approaches for targeting EOC. Unraveling the mechanisms underlying EOC metastasis will open up avenues to the design of new therapeutic options. For instance, understanding the molecular mechanisms involved in the hematogenous metastasis of EOC, the biology of CTCs, and the detailed mechanisms through which EOC cells take advantage of stromal cells may help to find new opportunities for targeting EOC metastasis.


Asunto(s)
Carcinoma Epitelial de Ovario/patología , Metástasis de la Neoplasia/patología , Neoplasias Ováricas/patología , Femenino , Humanos
7.
Cell Oncol (Dordr) ; 43(1): 31-49, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31828552

RESUMEN

BACKGROUND: Lung cancer is the second most common cancer and the main cause of cancer-related mortality worldwide. In spite of various efforts that have been made to facilitate the early diagnosis of lung cancer, most patients are diagnosed when the disease is already in stage IV, which is generally associated with the occurrence of distant metastases and a poor survival. Moreover, a large proportion of these patients will relapse after treatment, heralding the need for the stratification of lung cancer patients in addition to identifying those who are at a higher risk of relapse and, thus, require alternative and/or additional therapies. Recently, circulating tumor cells (CTCs) have been considered as valuable markers for the early diagnosis, prognosis and risk stratification of cancer patients, and they have been found to be able to predict the survival of patients with various types of cancer, including lung cancer. Additionally, the characterization of CTCs has recently provided fascinating insights into the heterogeneity of tumors, which may be instrumental for the development of novel targeted therapies. CONCLUSIONS: Here we review our current understanding of the significance of CTCs in lung cancer metastasis. We also discuss prominent studies reporting the utility of enumeration and characterization of CTCs in lung cancer patients as prognostic and pharmacodynamic biomarkers for those who are at a higher risk of metastasis and drug resistance.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Células Neoplásicas Circulantes/inmunología , Células Neoplásicas Circulantes/patología , Biomarcadores de Tumor/inmunología , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal/inmunología , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Células Neoplásicas Circulantes/metabolismo , Pronóstico
8.
Biosens Bioelectron ; 130: 1-19, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30716589

RESUMEN

Mucin 1 protein (MUC1) is a membrane-associated glycoprotein overexpressed in the majority of human malignancies and considered as a predominant protein biomarker in cancers. Owing to the crucial role of MUC1 in cancer dissemination and metastasis, detection and quantification of this biomarker is of great importance in clinical diagnostics. Today, there exist a wide variety of strategies for the determination of various types of disease biomarkers, especially MUC1. In this regard, aptamers, as artificial single-stranded DNA or RNA oligonucleotides with catalytic and receptor properties, have drawn lots of attention for the development of biosensing platforms. So far, various sensitivity-enhancement techniques in combination with a broad range of smart nanomaterials have integrated into the design of novel aptamer-based biosensors (aptasensors) to improve detection limit and sensitivity of analyte determination. This review article provides a brief classification and description of the research progresses of aptamer-based biosensors and nanobiosensors for the detection and quantitative determination of MUC1 based on optical and electrochemical platforms.


Asunto(s)
Técnicas Biosensibles , Técnicas Electroquímicas , Mucina-1/aislamiento & purificación , Neoplasias/diagnóstico , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/genética , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , Humanos , Mucina-1/química , Mucina-1/genética , Nanoestructuras/química
9.
Cell Oncol (Dordr) ; 41(2): 123-140, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29568985

RESUMEN

BACKGROUND: Breast cancer (BC) is the most common type of cancer in women and the second cause of cancer-related mortality world-wide. The majority of BC-related deaths is due to metastasis. Bone, lung, brain and liver are the primary target sites of BC metastasis. The clinical implications and mechanisms underlying bone metastasis have been reviewed before. Given the fact that BC lung metastasis (BCLM) usually produces symptoms only after the lungs have been vastly occupied with metastatic tumor masses, it is of paramount importance for diagnostic and prognostic, as well as therapeutic purposes to comprehend the molecular and cellular mechanisms underlying BCLM. Here, we review current insights into the organ-specificity of BC metastasis, including the role of cancer stem cells in triggering BC spread, the traveling of tumor cells in the blood stream and their migration across endothelial barriers, their adaptation to the lung microenvironment and the initiation of metastatic colonization within the lung. CONCLUSIONS: Detailed understanding of the mechanisms underlying BCLM will shed a new light on the identification of novel molecular targets to impede daunting pulmonary metastases in patients with breast cancer.


Asunto(s)
Neoplasias de la Mama/complicaciones , Metástasis de la Neoplasia/patología , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Femenino , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario
10.
Biosens Bioelectron ; 110: 23-37, 2018 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-29579646

RESUMEN

Vascular endothelial growth factor (VEGF) is a key regulator of vascular formation and a predominant protein biomarker in cancer angiogenesis. Owing to its crucial roles in the cancer metastasis, VEGF detection and quantification is of great importance in clinical diagnostics. Today, there exist a wide variety of detection strategies for identifying many types of disease biomarkers, especially for VEGF. As artificial single-stranded DNA or RNA oligonucleotides with catalytic and receptor properties, aptamers have drawn lots of attention to be applied in biosensing platforms due to their target-induced conformational changes as well as high stability and target versatility. So far, various sensitivity-enhancement techniques in combination with a broad range of smart nanomaterials have integrated into the design of novel aptasensors to improve detection limit and sensitivity of analyte detection. This review article provides a brief classification and description of the research progresses of aptamer-based biosensors and nanobiosensors for the detection and quantitative determination of VEGF based on optical and electrochemical platforms.


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Nanoestructuras/química , Factor A de Crecimiento Endotelial Vascular/análisis , Animales , Biomarcadores de Tumor/análisis , Técnicas Biosensibles/instrumentación , ADN de Cadena Simple/química , Técnicas Electroquímicas/instrumentación , Técnicas Electroquímicas/métodos , Diseño de Equipo , Humanos , Neoplasias/diagnóstico
11.
Int J Hematol Oncol Stem Cell Res ; 11(3): 250-261, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28989593

RESUMEN

Colorectal cancer (CRC) is one of the most common cancers worldwide and considered to be one of the hassles in medical communities. CRC develops from precancerous polyps in the colon or rectum and is preventable and curable by an early diagnosis and with the removal of premalignant polyps. In recent years, scientists have looked for inexpensive and safe ways to detect CRC in its earliest stages. Strong evidence shows that screening for CRC is a crucial way to reduce the incidence and mortality of this devastating disease. The main purpose for screening is to detect cancer or pre-cancer signs in all asymptomatic patients. In this review, we holistically introduce major pathways involved in the initiation and progression of colorectal tumorgenesis, which mainly includes chromosome instability (CIN), microsatellite instability (MSI), the CpG island methylator phenotype (CIMP), and we then will discuss different screening tests and especially the latest non-invasive fecal screening test kits for the detection of CRC.

12.
Cell Oncol (Dordr) ; 40(5): 419-441, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28921309

RESUMEN

BACKGROUND: Lung cancer is the most common cause of cancer-related mortality in humans. There are several reasons for this high rate of mortality, including metastasis to several organs, especially the brain. In fact, lung cancer is responsible for approximately 50% of all brain metastases, which are very difficult to manage. Understanding the cellular and molecular mechanisms underlying lung cancer-associated brain metastasis brings up novel therapeutic promises with the hope to ameliorate the severity of the disease. Here, we provide an overview of the molecular mechanisms underlying the pathogenesis of lung cancer dissemination and metastasis to the brain, as well as promising horizons for impeding lung cancer brain metastasis, including the role of cancer stem cells, the blood-brain barrier, interactions of lung cancer cells with the brain microenvironment and lung cancer-driven systemic processes, as well as the role of growth factor/receptor tyrosine kinases, cell adhesion molecules and non-coding RNAs. In addition, we provide an overview of current and novel therapeutic approaches, including radiotherapy, surgery and stereotactic radiosurgery, chemotherapy, as also targeted cancer stem cell and epithelial-mesenchymal transition (EMT)-based therapies, micro-RNA-based therapies and other small molecule or antibody-based therapies. We will also discuss the daunting potential of some combined therapies. CONCLUSIONS: The identification of molecular mechanisms underlying lung cancer metastasis has opened up new avenues towards their eradication and provides interesting opportunities for future research aimed at the development of novel targeted therapies.


Asunto(s)
Neoplasias Encefálicas/secundario , Transición Epitelial-Mesenquimal , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Quimioterapia/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , MicroARNs/genética , Células Madre Neoplásicas/metabolismo , Radioterapia/métodos , Procedimientos Quirúrgicos Operativos/métodos
13.
J Immunotoxicol ; 14(1): 137-151, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28693405

RESUMEN

Behçet's disease (BD) is a chronic autoimmune condition primarily prevalent in populations along the Mediterranean Sea. The exact etiology of BD has not been fully explained yet, but the disease occurrence is associated with a genetic factor, human leukocyte antigen (HLA)-B51 antigen. Among the various immunodysfunctions that are found in BD, patients are increased neutrophil motility and superoxide production, as well as elevated production of tumor necrosis factor (TNF)-α and decreased production of interleukin (IL)-10. Elevated levels of inflammatory cytokines like IL-1 and IL-17 in BD have been found associated with aberrant expression of microRNA. Gene polymorphisms in BD patients have been observed in molecules involved in responses to pathogens that can ultimately modulate the host antimicrobial response. Moreover, several single nucleotide polymorphisms (SNPs) have been reported in genes encoding chemokines and adhesion molecules; many of these changes manifest as increases in vascular inflammation and vascular damage. Lastly, genetic and epigenetic changes have been suggested as involved in the pathogenesis of BD. Modifications in DNA methylation have been found in BD patient monocytes and lymphocytes, leading to adverse function of these cells. This review presents a comprehensive compilation of the literature with regard to the immunodysfunction underlying BD, as well as of the genetics, newly described clinical specifications and novel treatment strategies using immunomodulants based on the current understanding of BD.


Asunto(s)
Síndrome de Behçet/genética , Síndrome de Behçet/inmunología , Antígeno HLA-B51/genética , MicroARNs/genética , Neutrófilos/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Movimiento Celular , Metilación de ADN , Epigénesis Genética , Predisposición Genética a la Enfermedad , Humanos , Inmunomodulación , Interleucina-1/metabolismo , Interleucina-17/metabolismo , Polimorfismo de Nucleótido Simple , Superóxidos/metabolismo
14.
Asian Pac J Cancer Prev ; 17(3): 1553-64, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27039805

RESUMEN

Identification of novel therapeutics in glioblastoma remains crucial due to the devastating and infiltrative capacity of this malignancy. The current study was aimed to appraise effect of arsenic trioxide (ATO) in U87MG cells. The results demonstrated that ATO induced apoptosis and impeded proliferation of U87MG cells in a dosedependent manner and also inhibited classical NF-κB signaling pathway. ATO further upregulated expression of Bax as an important proapoptotic target of NF-κB and also inhibited mRNA expression of survivin, c-Myc and hTERT and suppressed telomerase activity. Moreover, ATO significantly increased adhesion of U87MG cells and also diminished transcription of NF-κB down-stream targets involved in cell migration and invasion, including cathepsin B, uPA, MMP-2, MMP-9 and MMP-14 and suppressed proteolytic activity of cathepsin B, MMP-2 and MMP-9, demonstrating a possible mechanism of ATO effect on a well-known signaling in glioblastoma dissemination. Taken together, here we suggest that ATO inhibits survival and invasion of U87MG cells possibly through NF-κB-mediated inhibition of survivin and telomerase activity and NF-κB-dependent suppression of cathepsin B, MMP-2 and MMP-9.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Proliferación Celular/efectos de los fármacos , Glioblastoma/patología , FN-kappa B/metabolismo , Óxidos/farmacología , Trióxido de Arsénico , Western Blotting , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , FN-kappa B/genética , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Telomerasa/genética , Telomerasa/metabolismo , Células Tumorales Cultivadas
15.
Ann Hematol ; 95(7): 1031-42, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27091351

RESUMEN

Aurora B kinase as a chromosomal passenger protein plays multiple roles in regulating mitosis and cytokinesis. The function of Aurora B in leukemic cells has made it an important treatment target. In this study, we explored the expressions of Aurora (A, B, and C) kinases in newly diagnosed acute promyelocytic leukemia (APL) patients. In addition, we investigated the effects of AZD1152 as a specific inhibitor of Aurora B on cell survival, DNA synthesis, nuclear morphology, apoptosis induction, cell cycle distribution, and gene expression in an APL-derived NB4 cell line. Our results showed that Aurora B was overexpressed in 88 % of APL patients. AZD1152 treatment of NB4 cells led to viability reduction and G2/M arrest followed by an increase in cell size and polyploidy induction. These giant cells showed morphological evidence of mitotic catastrophe. AZD1152 treatment induced activation of G2/M checkpoint which in turn led to transient G2/M arrest in a p21-independent manner. Lack of functional p53 in NB4 cells might provide an opportunity to escape from G2/M block and to endure repeated rounds of replication and polyploidy. Treated cells were probably eliminated via p73-mediated overexpression of BAX, PUMA, and APAF1 and downregulation of survivin and MCL-1. In summary, AZD1152 treatment led to endomitosis and polyploidy in TP53-mutated NB4 cells. These giant polyploid cells might undergo mitotic catastrophe and p73-mediated apoptosis. It seems that induction of polyploidy via AZD1152 could be a novel form of anti-cancer therapy for APL that may be clinically accessible in the near future.


Asunto(s)
Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa B/biosíntesis , Regulación Neoplásica de la Expresión Génica , Leucemia Promielocítica Aguda/tratamiento farmacológico , Organofosfatos/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Adulto , Aurora Quinasa B/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Masculino , Persona de Mediana Edad , Organofosfatos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Resultado del Tratamiento
16.
Arch Iran Med ; 17(5): 366-71, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24784867

RESUMEN

BACKGROUND: Silibinin is a traditionally well-known drug for its hepatoprotective efficacy against various types of liver afflictions. In addition, it has recently been considered broadly as a potential chemopreventive agent against many types of cancers. The current study was designed to evaluate the restrictive effects of pharmacological doses of silibinin on SKBR3, an ErbB2-overexpressed and ER-negative human breast carcinoma cell line. METHODS: Effect of silibinin on metabolic activity and proliferation of human breast carcinoma (SKBR3) cell line were evaluated by MTT and BrdU assays respectively. Furthermore, the proapoptotic effect of silibinin was investigated using flow cytometry. The NF-κB phosphorylation assay was also used to assess the effect of silibinin on NF-κB activation. The alkalizing effect of silibinin on SKBR3 cell line was evaluated by measuring pH of media of the silibinin-treated cells compared to control. RESULTS: Our results indicate that silibinin inhibited metabolic activity and cell proliferation of SKBR3 cells in a dose-dependent manner. Moreover, silibinin significantly induced apoptosis in SKBR3 cells. On the other hand, silibinin significantly inhibited activation of NF-κB which is known to be highly active in this cell line. Alkalizing effect of silibinin was also observed. CONCLUSION: The results obtained here indicate that silibinin may be an efficacious therapeutic agent against ER-negative breast carcinomas with high inhibitory effect on NF-κB.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Silimarina/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN de Neoplasias/biosíntesis , Relación Dosis-Respuesta a Droga , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Fosforilación/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Silibina , Silimarina/uso terapéutico
17.
Neurochem Int ; 63(8): 809-17, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24161621

RESUMEN

PI3K/Akt and MAPK/ERK pathways are differentially activated in neuroblastoma (NB) cell types. In an effort to enhance the effectiveness of the NB treatment, we designed experiments to evaluate the effects of ATO in combination with PI3K and MEK1/2 specific inhibitors, LY29004 and U0126, respectively, in SK-N-MC and SK-N-BE(2) cell lines. The results indicated that specific inhibition of PI3K and MEK1/2 significantly enhanced antiproliferative and proapoptotic effects of ATO in SK-N-BE(2), but not in SK-N-MC. Furthermore, in SK-N-BE(2), NF-κB activation was significantly suppressed by LY29004+ATO treatments as compared with ATO alone, indicating that inhibition of PI3K may enhance anti-neoplastic properties of ATO in I-type NB cells through suppression of NF-κB. Moreover, expressions of c-Myc, Bad, Bax and ATM in SK-N-BE(2) cell line were significantly increased by U0126+ATO treatment as compared to treatment with ATO alone. Expression of telomerase hTERT was almost depleted by U0126+ATO treatment. Regarding the fact that activation of PI3K and MAPK in SK-N-BE(2) is higher than in other NB subtypes, we hypothesize that growth of SK-N-BE(2) cell line is highly dependent on these pathways and inhibition of these pathways may has promise for the treatment of multi-drug resistant I-type NB cells by ATO. However, for successful strategies for the treatment of this heterogeneous tumor, other combinations approaches need to be considered to simultaneously target other NB cells.


Asunto(s)
Arsenicales/farmacología , Neuroblastoma/patología , Óxidos/farmacología , Proteínas Quinasas/metabolismo , Apoptosis , Trióxido de Arsénico , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Cartilla de ADN , Activación Enzimática , Citometría de Flujo , Humanos , Neuroblastoma/enzimología , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
Mol Cell Endocrinol ; 377(1-2): 84-92, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23851143

RESUMEN

Human growth hormone (hGH) has been increasingly implicated in a variety of cancers; its up-regulation is observed in breast cancer and correlates with a poor outcome. Autocrine hGH promotes mammary carcinoma cell survival, proliferation, immortalization; it confers an invasive phenotype as a result of an epithelial-mesenchymal transition and contributes to chemoresistance and radioresistance. Arsenic trioxide (ATO) is being successfully used as a first and second line therapy for the treatment of patients with acute promyelocytic leukemia. It also inhibits tumor cell growth and induces apoptosis in a broad range of solid tumors. In the present study, we investigated the effect of hGH on sensitivity of a mammary adenocarcinoma cell to ATO, using a stable hGH-transfectant MCF-7 cell line, MCF7-hGH. Our results demonstrated for the first time that the overexpression of hGH increased sensitivity of the breast cancer cell line MCF-7 to ATO through apoptotic and anti-proliferative mechanisms. The effect of ATO on the transcriptional level of genes involved in survival (Bcl-2, Bax and Survivin), self-sufficiency in growth signals (c-Myc, ARF, Cdc25A, p53 and Bax), immortalization (hTERT) and invasion and metastasis (MMP-2 and MMP-9, uPA and uPAR and E-cadherin) was more pronounced in MCF7-hGH compared with its parental MCF-7 line. Our study may highlight the potential application of ATO for the treatment of patients with breast cancer, especially in those who have metastatic and chemoresistant tumor phenotype possibly due to the over expression of hGH.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Comunicación Autocrina/efectos de los fármacos , Neoplasias de la Mama/patología , Hormona de Crecimiento Humana/metabolismo , Óxidos/farmacología , Trióxido de Arsénico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Metaloproteinasas de la Matriz/metabolismo , Modelos Biológicos , Metástasis de la Neoplasia , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Ensayo de Tumor de Célula Madre , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
19.
Ann Hematol ; 92(9): 1207-20, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23645216

RESUMEN

To enhance anticancer efficacy of the arsenic trioxide (ATO), the combination of ATO and azidothymidine (AZT), with convergence anti-telomerase activity, were examined on acute promyelocytic leukemia (APL) cell line, NB4. In spite of an induction of apoptosis by both drugs separately and a synergistic effect of them on hTERT down-regulation and telomerase inhibition, the ATO-induced cytotoxicity was reduced when it was used in combination with AZT. AZT attenuated the ATO effects on viability, metabolic activity, DNA synthesis, and apoptosis. These observations, despite the deflection from the main goal of this study, dedicate an especial opportunity to elucidate the importance of some of the mechanisms that have been suggested by which ATO induces apoptosis. Cell cycle distribution, ROS level, and caspase-3 activation analyses suggest that AZT reduced the ATO-induced cytotoxic effect possibly via relative induction and diminution of cells accumulated in (G1, S) and (G2/M) phase, respectively, as well as through attenuation of ROS generation and subsequent caspase-3 inhibition. QRT-PCR assay revealed that induction of p21expression by the combined AZT/ATO compared to ATO alone could be a reason for the relative decline of cells accumulation in G2/M and the increase of cells in G1 and S phases. Therefore, the G2/M arrest and ROS generation are likely principle mediators for the ATO-induced apoptosis and can be used as a guide to design rational combinatorial strategies involving ATO and agents with G2/M arrest or ROS generation capacity to intensify ATO-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Leucemia Promielocítica Aguda/patología , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Óxidos/toxicidad , Zidovudina/uso terapéutico , Apoptosis/fisiología , Trióxido de Arsénico , Arsenicales/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Óxidos/antagonistas & inhibidores
20.
Neurochem Res ; 37(9): 2053-63, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22717697

RESUMEN

Neuroblastoma is the most common solid tumor in children. Current therapy modalities have resulted in little amelioration in the cure rate of neuroblsatoma and therefore, outlining biologically based therapies for neuroblastoma remains of main priority. This study was carried out to appraise the impeding effects of silibinin, a potent anti-cancer agent, on two different neuroblastoma cell lines, stromal SK-N-MC and neuroblastic SK-N-BE(2) cells. The microculture tetrazolium assay, gelatin zymography, colony formation assay, cell cycle distribution survey, apoptosis assay, and quantitative real-time reverse transcription-PCR were applied to evaluate the effects of silibinin on metabolic activity, gelatinolytic activity of MMP-2 and MMP-9, surviving potential, cell cycle, apoptosis, and expression pattern of the genes involved in cell survival and invasion of the two neuroblastoma cell lines. Treatment for 48 h inhibited metabolic activity and clonogenic potential of SK-N-MC cells in a dose-dependent manner. Silibinin also inhibited transcriptional levels of MMP-2, MMP-9, and uPAR, as markers of cell invasion, in SK-N-MC cells. Higher concentration of silibinin (75, 100 µM) suppressed enzymatic activity of MMP-2 in this cell line. No change in apoptosis and cell cycle was observed in neither of the cells after treatment with silibinin. On the other hand, silibinin highly decreased mRNA expression of Akt, and NF-κB1 and its regulators, IKK1 and IKK2 in SK-N-MC cell line. Comparison of transcriptional expression of Akt, and NF-κB1 in untreated stromal and neuroblastic cell lines shows that their basal transcriptional levels are much higher in SK-N-BE(2) cell line than that in SK-N-MC cells. It seems that SK-N-BE(2) cell line probably resists to silibinin through higher expression of Akt and probably NF-κB1. Collectively, our results demonstrated that silibinin highly inhibits the proliferative potentials of SK-N-MC cell line, whilst it had less inhibitory effect on SK-N-BE(2) cell line. Our results suggest that suppression of SK-N-MC cell line by silibinin may be through inhibition of Akt-mediated NF-κB1.


Asunto(s)
Antineoplásicos , FN-kappa B/biosíntesis , Neuroblastoma/tratamiento farmacológico , Proteína Oncogénica v-akt/biosíntesis , Sustancias Protectoras/farmacología , Silimarina/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Dosificación Letal Mediana , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , FN-kappa B/genética , Neuroblastoma/patología , Proteína Oncogénica v-akt/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Silibina , Sales de Tetrazolio , Tiazoles , Transcripción Genética/efectos de los fármacos , Ensayo de Tumor de Célula Madre , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA