Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cancer Lett ; 591: 216891, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38642607

RESUMEN

Ovarian cancer ranks as a leading cause of mortality among gynecological malignancies, primarily due to the lack of early diagnostic tools, effective targeted therapy, and clear understanding of disease etiology. Previous studies have identified the pivotal role of Lysophosphatidic acid (LPA)-signaling in ovarian cancer pathobiology. Our earlier transcriptomic analysis identified Urothelial Carcinoma Associated-1 (UCA1) as an LPA-stimulated long non-coding RNA (lncRNA). In this study, we elucidate the tripartite interaction between LPA-signaling, UCA1, and let-7 miRNAs in ovarian cancer progression. Results show that the elevated expression of UCA1 enhances cell proliferation, invasive migration, and therapy resistance in high-grade serous ovarian carcinoma cells, whereas silencing UCA1 reverses these oncogenic phenotypes. UCA1 expression inversely correlates with survival outcomes and therapy response in ovarian cancer clinical samples, underscoring its prognostic significance. Mechanistically, UCA1 sequesters let-7 miRNAs, effectively neutralizing their tumor-suppressive functions involving key oncogenes such as Ras and c-Myc. More significantly, intratumoral delivery of UCA1-specific siRNAs inhibits the growth of cisplatin-refractory ovarian cancer xenografts, demonstrating the therapeutic potential of targeting LPAR-UCA1-let-7 axis in ovarian cancer. Thus, our results identify LPAR-UCA1-let-7 axis as a novel avenue for targeted treatment strategies.


Asunto(s)
Movimiento Celular , Proliferación Celular , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , MicroARNs , Neoplasias Ováricas , ARN Largo no Codificante , Femenino , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , MicroARNs/genética , MicroARNs/metabolismo , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Transducción de Señal , Ratones Desnudos , Lisofosfolípidos/metabolismo , Ratones , Cisplatino/farmacología , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo
2.
Metabolites ; 13(5)2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-37233659

RESUMEN

Peritoneal cancers present significant clinical challenges with poor prognosis. Understanding the role of cancer cell metabolism and cancer-promoting metabolites in peritoneal cancers can provide new insights into the mechanisms that drive tumor progression and can identify novel therapeutic targets and biomarkers for early detection, prognosis, and treatment response. Cancer cells dynamically reprogram their metabolism to facilitate tumor growth and overcome metabolic stress, with cancer-promoting metabolites such as kynurenines, lactate, and sphingosine-1-phosphate promoting cell proliferation, angiogenesis, and immune evasion. Targeting cancer-promoting metabolites could also lead to the development of effective combinatorial and adjuvant therapies involving metabolic inhibitors for the treatment of peritoneal cancers. With the observed metabolomic heterogeneity in cancer patients, defining peritoneal cancer metabolome and cancer-promoting metabolites holds great promise for improving outcomes for patients with peritoneal tumors and advancing the field of precision cancer medicine. This review provides an overview of the metabolic signatures of peritoneal cancer cells, explores the role of cancer-promoting metabolites as potential therapeutic targets, and discusses the implications for advancing precision cancer medicine in peritoneal cancers.

3.
Cells ; 11(16)2022 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-36010595

RESUMEN

The cellular signaling network involves co-ordinated regulation of numerous signaling molecules that aid the maintenance of cellular as well as organismal homeostasis. Aberrant signaling plays a major role in the pathophysiology of many diseases. Recent studies have unraveled the superfamily of long non-coding RNAs (lncRNAs) as critical signaling nodes in diverse signaling networks. Defective signaling by lncRNAs is emerging as a causative factor underlying the pathophysiology of many diseases. LncRNAs have been shown to be involved in the multiplexed regulation of diverse pathways through both genetic and epigenetic mechanisms. They can serve as decoys, guides, scaffolds, and effector molecules to regulate cell signaling. In comparison with the other classes of RNAs, lncRNAs possess unique structural modifications that contribute to their diversity in modes of action within the nucleus and cytoplasm. In this review, we summarize the structure and function of lncRNAs as well as their vivid mechanisms of action. Further, we provide insights into the role of lncRNAs in the pathogenesis of four major disease paradigms, namely cardiovascular diseases, neurological disorders, cancers, and the metabolic disease, diabetes mellitus. This review serves as a succinct treatise that could open windows to investigate the role of lncRNAs as novel therapeutic targets.


Asunto(s)
ARN Largo no Codificante , Núcleo Celular , Epigénesis Genética , Homeostasis , ARN Largo no Codificante/genética , Transducción de Señal
4.
Am J Cancer Res ; 12(1): 17-47, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35141003

RESUMEN

BRCA1 mutation carriers have a greater risk of developing cancers in hormone-responsive tissues like breasts and ovaries. However, this tissue-specific incidence of BRCA1 related cancers remains elusive. The majority of the BRCA1 mutated breast cancers exhibit typical histopathological features of high-grade tumors, with basal epithelial phenotype, classified as triple-negative molecular subtype and have a higher percentage of DNA damage and chromosomal abnormality. Though there are many studies relating BRCA1 with ER-α (Estrogen receptor-α), it has not been reported whether E2 (Estrogen) -ER-α signaling can modulate the DNA repair activities of BRCA1. The present study analyzes whether deregulation of ER-α signaling, arising as a result of E2/ER-α deficiency, could impact the BRCA1 dependent DDR (DNA Damage Response) pathways, predominantly those of DNA-DSB (Double Strand break) repair and oxidative damage response. We demonstrate that E2/E2-stimulated ER-α can augment BRCA1 mediated high fidelity repairs like HRR (Homologous Recombination Repair) and BER (Base Excision Repair) in breast cancer cells. Conversely, a condition of ER-α deficiency itself or any interruption in ligand-dependent ER-α transactivation resulted in delayed DNA damage repair, leading to persistent activation of γH2AX and retention of unrepaired DNA lesions, thereby triggering tumor progression. ER-α deficiency not only limited the HRR in cells but also facilitated the DSB repair through error prone pathways like NHEJ (Non Homologous End Joining). ER-α deficiency associated persistence of DNA lesions and reduced expression of DDR proteins were validated in human mammary tumors.

5.
Biomedicines ; 9(12)2021 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-34944743

RESUMEN

Focusing on defining metabolite-based inter-tumoral heterogeneity in ovarian cancer, we investigated the metabolic diversity of a panel of high-grade serous ovarian carcinoma (HGSOC) cell-lines using a metabolomics platform that interrogate 731 compounds. Metabolic fingerprinting followed by 2-dimensional and 3-dimensional principal component analysis established the heterogeneity of the HGSOC cells by clustering them into five distinct metabolic groups compared to the fallopian tube epithelial cell line control. An overall increase in the metabolites associated with aerobic glycolysis and phospholipid metabolism were observed in the majority of the cancer cells. A preponderant increase in the levels of metabolites involved in trans-sulphuration and glutathione synthesis was also observed. More significantly, subsets of HGSOC cells showed an increase in the levels of 5-Hydroxytryptamine, γ-aminobutyrate, or glutamate. Additionally, 5-hydroxytryptamin synthesis inhibitor as well as antagonists of γ-aminobutyrate and glutamate receptors prohibited the proliferation of HGSOC cells, pointing to their potential roles as oncometabolites and ligands for receptor-mediated autocrine signaling in cancer cells. Consistent with this role, 5-Hydroxytryptamine synthesis inhibitor as well as receptor antagonists of γ-aminobutyrate and Glutamate-receptors inhibited the proliferation of HGSOC cells. These antagonists also inhibited the three-dimensional spheroid growth of TYKNU cells, a representative HGSOC cell-line. These results identify 5-HT, GABA, and Glutamate as putative oncometabolites in ovarian cancer metabolic sub-type and point to them as therapeutic targets in a metabolomic fingerprinting-based therapeutic strategy.

6.
Biochim Biophys Acta Rev Cancer ; 1875(1): 188482, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33260050

RESUMEN

Carriers of BRCA1 mutations have a higher chance of developing cancers in hormone-responsive tissues like the breast, ovary and prostate, compared to other tissues. These tumors generally exhibit basal-like characters and do not express estrogen receptor (ER) or progesterone receptor (PR). Intriguingly, BRCA1 mutated breast cancers have a less favorable clinical outcome, as they will not respond to hormone therapy. BRCA1 has been reported to exhibit ligand dependent and independent transcriptional inhibition of ER-α; however, there exists a controversy on whether BRCA1 induces or inhibits ER-α expression. The mechanisms associated with resistance of BRCA1 mutated cancers to hormone therapy, as well as the tissue restriction exhibited by BRCA1 mutated tumors are still largely unknown. BRCA1 mutated tumors possess increased DNA damages and decreased genomic integrity, as BRCA1 plays a cardinal role in high fidelity DNA damage repair pathways, like homologous recombination (HR). The existence of cross regulatory signaling networks between ER-α and BRCA1 speculates a role of ER on BRCA1 dependent DDR pathways. Thus, the loss or haploinsufficiency of BRCA1 and the consequential deregulation of ER-α signaling may result in persistence of unrepaired DNA damages, eventually leading to tumorigenesis. Therefore, understanding of this cross-talk between ER-α and BRCA1, with regard to DDR, will provide critical insights to steer drug development and therapy for breast/ovarian cancers. This review discusses the mechanisms by which estrogen and ER signaling influence BRCA1 mediated DNA damage response and repair pathways in the mammalian system.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Carcinogénesis/genética , Receptor alfa de Estrógeno/genética , Mama/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Femenino , Humanos , Transducción de Señal/efectos de los fármacos
7.
Adv Cancer Res ; 147: 319-373, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32593405

RESUMEN

Non-communicable diseases contribute to 71% of the deaths worldwide, of which cancers rank second after cardiovascular diseases. Among all the cancers, head and neck cancers (HNC) are consequential in augmenting the global cancer incidence as well as mortality. Receptor tyrosine kinases (RTKs) are emphatic for the matter that they serve as biomarkers aiding the analysis of tumor progression and metastasis as well as diagnosis, prognosis and therapeutic progression in the patients. The extensive researches on HNC have made significant furtherance in numerous targeted therapies, but for the escalating therapeutic resistance. This review explicates RTKs in HNC, their signaling pathways involved in tumorigenesis, metastasis and stemness induction, the association of non-coding RNAs with RTKs, an overview of RTK based therapy and associated resistance in HNC, as well as a sneak peek into the HPV positive HNC and its therapy. The review extrapolates the cardinal role of RTKs and RTK based therapy as superior to other existing therapeutic interventions for HNC.


Asunto(s)
Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/enzimología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Animales , Neoplasias de Cabeza y Cuello/patología , Humanos , Terapia Molecular Dirigida/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal
8.
Crit Rev Oncol Hematol ; 151: 102964, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32464482

RESUMEN

Breast Cancer is the most predominant female cancer in developed as well as developing countries. The treatment strategies of breast cancers depends on an array of factors like age at diagnosis, menstrual status, dietary pattern, immunological response, genetic variations of the cancer cells etc. Recent technological advancements in cancer diagnosis lead to the emergence of gene expression pattern for better understanding of the tumor behavior. It has not only bolstered the prognosis, but also the early diagnosis and therapy. The accuracy in disease prognosis can be boosted when gene expression signatures are combined with traditional clinicopathological features. This review explains how the evolution of gene expression signatures for breast cancers, its advantages and future prospects. In addition, an overview of currently available gene expression signature analysis tools and consolidated information on their current status and specific benefits, that can be availed for breast cancer diagnosis are also discussed.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Neoplasias de la Mama/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Pronóstico , Transcriptoma
9.
Carcinogenesis ; 41(5): 611-624, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31219560

RESUMEN

Gestational trophoblastic diseases (GTD) are group of pregnancy-related tumors characterized by abnormal levels of 'ß-hCG' with higher incidence in South-East Asia, especially India. Our laboratory has reported that wild-type BRCA1 transcriptionally regulates ß-hCG in triple negative breast cancers (TNBCs). These factors culminated into analysis of BRCA1 status in GTD, which would emanate into elucidation of BRCA1- ß-hCG relationship and unraveling etio-pathology of GTD. BRCA1 level in GTD is down-regulated due to the over-expression of DNMT3b and subsequent promoter hypermethylation, when compared to the normal placentae accompanied with its shift in localization. There is an inverse correlation of serum ß-hCG levels with BRCA1 mRNA expression. The effects of methotrexate (MTX), which is the first-line chemotherapeutic used for GTD treatment, when analyzed in comparison with plumbagin (PB) revealed that PB alone is efficient than MTX alone or MTX-PB in combination, in showing selective cytotoxicity against GTD. Interestingly, PB increases BRCA1 levels post-treatment, altering DNMT3b levels and resultant BRCA1 promoter methylation. Also, cohort study analyzed the incidence of GTD at Sree Avittom Thirunal (SAT) Hospital, Thiruvananthapuram, which points out that 11.5% of gestational trophoblastic neoplasia (GTN) cases were referred to Regional Cancer Centre, Thiruvananthapuram, for examination of breast lumps. This has lend clues to supervene the risk of GTD patients towards BRCA1-associated diseases and unveil novel therapeutic for GTD, a plant-derived naphthoquinone, PB, already reported as selectively cytotoxic against BRCA1 defective tumors.


Asunto(s)
Proteína BRCA1/genética , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Metilación de ADN , Enfermedad Trofoblástica Gestacional/patología , Mutación , Placenta/metabolismo , Regiones Promotoras Genéticas , Adulto , Antineoplásicos/farmacología , Apoptosis , Proteína BRCA2/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Gonadotropina Coriónica Humana de Subunidad beta/genética , Estudios de Cohortes , Femenino , Regulación Neoplásica de la Expresión Génica , Enfermedad Trofoblástica Gestacional/tratamiento farmacológico , Enfermedad Trofoblástica Gestacional/genética , Enfermedad Trofoblástica Gestacional/metabolismo , Humanos , Placenta/efectos de los fármacos , Placenta/patología , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/patología , Pronóstico , Neoplasias Trofoblásticas/tratamiento farmacológico , Neoplasias Trofoblásticas/genética , Neoplasias Trofoblásticas/metabolismo , Neoplasias Trofoblásticas/patología , Células Tumorales Cultivadas
10.
Front Vet Sci ; 6: 283, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31508437

RESUMEN

Human breast cancers (HBCs) are one of the leading causes of global cancer death among women. Domesticated canines are the most affected domestic species with a prevalence rate of breast cancer more than three times in women. While the human cancer patients receive substantial diagnostic and treatment facilities, inadequacy in canine cancer care, calls for greater attention. Fine Needle Aspiration Cytology (FNAC) is comparatively simple, quick, and easily reproducible technique, which aids in pre-surgical diagnosis. In humans, FNAC has a standard protocol, the Robinson's grading system, which has high correlation with the established histological grading system of Scarff Bloom- Richardson. However, Canine Mammary Tumors (CMTs), which are known to be similar to HBCs in biological behavior and gene expressions, still bank on the histopathological methods for diagnostic purposes. This review sheds light on various factors that could be considered for developing a standard FNAC technique for CMT grading and analyzes its future perspectives.

11.
Carcinogenesis ; 40(11): 1415-1426, 2019 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-30963174

RESUMEN

ß-hCG expression in breast cancer is highly controversial with reports supporting both protective and tumorigenic effects. It has also been reported that risk of breast cancer at an early age is increased with full-term pregnancies if a woman is a BRCA1 mutation carrier. We have already demonstrated that BRCA1-defective cells express high levels of ß-hCG and that when BRCA1 is restored, ß-hCG level is reduced. Also, BRCA1 can bind to the promoter and reduce the levels of ß-hCG. ß-hCG induces tumorigenicity in BRCA1-defective cells by directly binding to TGFBRII and induces TGFBRII-mediated cell proliferation. In this study, we analyzed the mechanism of action of ß-hCG on BRCA1 expression and its influence on drug sensitivity in breast cancer cells. We demonstrate that ß-hCG induces mutant BRCA1 protein expression in BRCA1 mutant cells; however, in BRCA1 wild-type cells, ß-hCG reduced wild-type BRCA1 protein expression. Transcriptionally, ß-hCG could induce Slug/LSD1-mediated repression of wild-type and mutant BRCA1 messenger RNA levels. However, ß-hCG induces HSP90-mediated stabilization of mutant BRCA1 and hence the overexpression of mutant BRCA1 protein, resulting in partial restoration of homologous recombination repair of damaged DNA. This contributes to drug resistance to HSP90 inhibitor 17AAG in BRCA1-defective cancer cells. A combination of HSP90 inhibitor and TGFBRII inhibitor has shown to sensitize ß-hCG expressing BRCA1-defective breast cancers to cell death. Targeting the ß-hCG-HSP90-TGFBRII axis could prove an effective treatment strategy for BRCA1-mutated breast tumors.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Gonadotropina Coriónica/metabolismo , Resistencia a Antineoplásicos , Animales , Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Gonadotropina Coriónica/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mutación , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Reparación del ADN por Recombinación , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Rep ; 8(1): 13903, 2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-30224826

RESUMEN

It is known that Cancer Associated Fibroblast (CAFs) from the primary tumor site can accompany cancer cells to a secondary site during the process of metastasis. We hypothesize that these CAFs could be transformed to an altered cell type, which can be called as Metastasis Associated Fibroblasts (MAF) in turn can support, and convoy cancer cells for metastasis. There are no published reports that have characterized and distinguished CAFs from MAF. It is well established that some of the cancer cells within the tumor mass accumulate novel mutations prior to metastasis. Hence, we speculated that mutations in the tumor suppressor gene, BRCA1, which is already reported to induce metastasis via abnormal expression of Ezrin, Radixin and Moesin (ERM), could generate MAF. In the present study, we demonstrate for the first time that CAFs isolated from primary breast cancer tissues when co-cultured with BRCA1 mutated HCC1937 cells transform CAFs to MAF in vitro. As expected, MAF augmented proliferation, migration and invasion along with over-expression of epithelial mesenchymal transition (EMT) markers, Ezrin and CCL5, thereby facilitating metastasis. Therefore, we inhibited Ezrin and CCL5 in vitro in MAF and observed that the migration and invasion abilities of these cells were attenuated. This highlights the intriguing possibilities of combination therapy using MAF inhibitors as anti-metastatic agents along with anticancer drugs, to control the metastatic spread from primary tumor site.


Asunto(s)
Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Genes BRCA1 , Metástasis de la Neoplasia , Neoplasias de la Mama/genética , Línea Celular Tumoral , Técnicas de Cocultivo , Transición Epitelial-Mesenquimal , Femenino , Fibroblastos/patología , Humanos
13.
Crit Rev Oncol Hematol ; 123: 74-82, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29482782

RESUMEN

Reports till its discovery has proven multiple facets of Breast Cancer type 1 susceptibility gene (BRCA1) from nucleus to cytoplasm; from DNA repair to drug resistance; from Homologous Recombination (HR) to Ubiquitination; from breast to brain; from cancer to HIV and many of the roles are still unexplored. One of the recent attractions of BRCA1 is its role in regulating breast cancer metastasis though the exact mechanism is poorly understood. In this review, we will discuss the molecular interactions between BRCA1 and the key molecules of Epithelial to Mesenchymal Transition (EMT) associated with metastasis, its associated drug resistance and the possible treatment strategy for BRCA1 mutated breast cancer.


Asunto(s)
Proteína BRCA1/fisiología , Neoplasias de la Mama/genética , Transición Epitelial-Mesenquimal/genética , Proteína BRCA1/genética , Neoplasias de la Mama/patología , Reparación del ADN/genética , Resistencia a Antineoplásicos/genética , Femenino , Recombinación Homóloga/genética , Humanos , Metástasis de la Neoplasia
14.
J Proteome Res ; 17(1): 276-289, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29028349

RESUMEN

Previously, we identified that ß-hCG is expressed by BRCA1 mutated but not wild type breast cancers in vitro/in vivo and exhibited a novel event in ß-hCG overexpressing BRCA1 mutated HCC1937 cells where the cells were able to form spheres (HCC1937 ß spheres) in adherent cell culture plates even in the absence of any growth factors. These spheres express stem cell and EMT markers. In the present study, we carried out the total proteomic profiling of these HCC1937 ß spheres obtained from BRCA1 defective ß-hCG expressing stable breast cancer cells to analyze the cell signaling pathways that are active in these cells. Functional annotation revealed proteins (164 cellular and 97 secretory) predominantly involved in oxygen binding, nucleosome assembly, cytoskeleton organization, protein folding, etc. Many of the proteins identified from HCC1937 ß spheres in this study are also up regulated in breast cancers, which are directly linked with poor prognosis in human cancer samples as analyzed using TCGA data set. Survival analysis shows that ß-hCG expressing cancer patients are linked with poor survival rate. Interestingly, hemoglobins were identified at both cellular and secretory level in HCC1937 ß spheres and experiments after treating with ROS inducers revealed that ß-hCG induces hemoglobin and protects the cancer cells during oxidative stress. Our proteomic data strongly propose ß-hCG as an oncogenic molecule associated with BRCA1 mutation, and hence, targeting ß-hCG could be a strategy to treat BRCA1 defective breast cancers.


Asunto(s)
Proteína BRCA1/genética , Gonadotropina Coriónica Humana de Subunidad beta/farmacología , Proteómica/métodos , Neoplasias de la Mama Triple Negativas/patología , Línea Celular Tumoral , Gonadotropina Coriónica Humana de Subunidad beta/uso terapéutico , Hemoglobinas/análisis , Humanos , Mutación , Estrés Oxidativo , Pronóstico , Análisis de Supervivencia
15.
Crit Rev Oncol Hematol ; 114: 77-90, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28477749

RESUMEN

Gestational trophoblastic diseases (GTD) encompass a group of placental tumors which mostly arise due to certain fertilization defects, resulting in the over-proliferation of trophoblasts. The major characteristic of this diseased state is that ß-hCG rises up manifold than that is observed during pregnancy. The incidence of GTD when analyzed on a global scale, figures out that there is a greater risk in South-East Asia, the reason of which remains unclear. An insight into any possible correlation of GTD incidence with cancers, other than choriocarcinoma, is being attempted here. Also, we review the recent developments in research on the molecular etiopathology of GTD. This review would render a wider eye towards a new paradigm of thoughts to connect GTD and breast cancer, which has not been into the picture till date.


Asunto(s)
Enfermedad Trofoblástica Gestacional/complicaciones , Neoplasias/etiología , Femenino , Enfermedad Trofoblástica Gestacional/epidemiología , Humanos , Incidencia , Neoplasias/epidemiología , Embarazo , Pronóstico
16.
Semin Oncol ; 44(4): 254-264, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-29526253

RESUMEN

Chromosome 17 (Chr17) harbors crucial genes that encode proteins implicated in a variety of cancers, including some that guard cancer cells from genomic instability and others that interfere with metastasis. Included amongst the genes on chr17 that regulate biological processes fundamental to the genesis of cancer are TP53, BRCA1, CCL5, NF-1, and GRB7. As many as 50% of all human tumors and at least 30% of breast carcinomas contain p53 mutations, while 30%-40% of breast cancers have defective BRCA1. A large number of proteins regulate the expression of these cancer genes on chr17 with miRNAs, the most widely studied class of regulatory RNAs, playing a major role in epigenetically controlling the gene expression programs, thereby managing various cellular functions. This review provides information on the genes transcribed from chr17, and their regulation by miRNAs in the context to tumorigenesis located on chr17, along with an analysis of the receptor status (estrogen, progesterone, and Her2/Neu) from the miRNA prediction data of miRNA genes located on chr17.


Asunto(s)
Cromosomas Humanos Par 17 , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Redes Reguladoras de Genes , Humanos
17.
Sci Rep ; 6: 26631, 2016 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-27220670

RESUMEN

We have earlier shown that Plumbagin (PB) can induce selective cytotoxicity to BRCA1 defective ovarian cancer cells; however, the effect of this molecule in BRCA1 mutated breast cancers has not been analyzed yet. Here, we report that reactive oxygen species (ROS) induced by PB resulted in DNA DSB and activates downstream signaling by ATR/ATM kinases and subsequent apoptosis. PB reduces DNA- dependent protein kinase (DNA-PK) expression and inhibits NHEJ (Non Homologous End Joining) activity in BRCA1 defective breast cancer cells. Also, PB induces apoptosis in two different BRCA1 conditional knock out murine models: MMTV-Cre; BRCA1(Co/Co) and WAP-Cre; BRCA1(Co/Co), at 2 mg/kg body weight, but 32 mg/kg of carboplatin (CN) was needed to induce apoptosis in them. This is the first study where two different tissue specific promoter driven transgenic mice models with BRCA1 exon 11 deletions are used for preclinical drug testing. The apoptosis induced by PB in HR (Homologous Recombination) defective triple negative BRCA1 mutant cell lines and in mouse models occur by inducing ROS mediated DNA DSB. The toxicity profile as compared with CN in transgenic mice provides evidence for PB's safer disposition as a therapeutic lead in breast cancer drug development.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína BRCA1/deficiencia , Roturas del ADN de Doble Cadena/efectos de los fármacos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Naftoquinonas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Proteínas Supresoras de Tumor/deficiencia , Animales , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
18.
Crit Rev Oncol Hematol ; 101: 184-92, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27017408

RESUMEN

The identification of various biomolecules in cancer progression and therapy has led to the exploration of the roles of two cardinal players, namely Nitric Oxide (NO) and Reactive Oxygen Species (ROS) in cancer. Both ROS and NO display bimodal fashions of functional activity in a concentration dependent manner, by inducing either pro- or anti- tumorigenic signals. Researchers have identified the potential capability of NO and ROS in therapies owing to their role in eliciting pro-apoptotic signals at higher concentrations and their ability to sensitize cancer cells to one another as well as to other therapeutics. We review the prospects of NO and ROS in cancer progression and therapy, and analyze the role of a combinatorial therapy wherein an NO donor (SNAP) is used to sensitize the oxidative damage repair defective, triple negative breast cancer cells (HCC 1937) to a potent ROS inducer. Preliminary findings support the potential to employ various combinatorial regimes for anti-cancer therapies with regard to exploiting the chemo-sensitization property of NO donors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Humanos , Células Madre Neoplásicas/metabolismo , Oxidación-Reducción
19.
Pharmacol Res ; 105: 134-45, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26808083

RESUMEN

Eventhough the role of BRCA1/2 in hereditary prostatic cancer is being unleashed at a rapid rate; their optimal clinical management remains undefined. Cancer stem cells are thought to be responsible for cancer chemoresistance and relapse, thus they represent a significant concern for cancer prognosis and therapy. In this study, we have analyzed the effect of Plumbagin (PB) and structurally related naphthaquinones on BRCA1/2 silenced prostate cancer cells and the ability of PB to target stem cells. Our cell proliferation studies showed that both PC-3 and DU145 cells were more sensitive to PB, though all the compounds induced mitochondrial potential loss, DNA fragmentation and morphological changes which are indicative of apoptosis. Both BRCA1/2 siRNA transfected PC-3 and DU145 cells exhibited increased sensitivity to PB. Gene expression profiling post PB treatment in BRCA1/2 silenced cells revealed that PB has a putative role in tumor suppression in BRCA defective cancers. Using flow cytometric analysis we have proved that PB has the putative ability to directly target CSCs. Overall studies suggest that PB's antitumour mechanisms holds promise for novel therapeutic approaches against BRCA mutated cancers as well as CSCs.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Proteína BRCA1/genética , Proteína BRCA2/genética , Naftoquinonas/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Próstata/efectos de los fármacos , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA