Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
NPJ Precis Oncol ; 7(1): 20, 2023 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-36801948

RESUMEN

ACE2 overexpression in colorectal cancer patients might increase susceptibility to SARS-CoV-2 infection. We report that knockdown, forced overexpression, and pharmacologic inhibition in human colon cancer cells targeted ACE2-BRD4 crosstalk to mediate marked changes in DNA damage/repair and apoptosis. In colorectal cancer patients for whom high ACE2 plus high BRD4 expression is predictive of poor survival, pan-BET inhibition would need to consider proviral/antiviral actions of different BET proteins during SARS-CoV-2 infection.

2.
Nutrients ; 14(20)2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36297001

RESUMEN

Epigenetic mechanisms play an important role in the etiology of colorectal cancer (CRC) and other malignancies due, in part, to deregulated bromodomain (BRD) functions. Inhibitors of the bromodomain and extraterminal (BET) family have entered into clinical trials as anticancer agents, and interest has grown in other acetyl 'reader' proteins as therapeutic targets, including non-BET member bromodomain-containing protein 9 (BRD9). We report here that overexpression of BRD9 is associated with poor prognosis in CRC patients, and that siRNA-mediated knockdown of BRD9 decreased cell viability and activated apoptosis in human colon cancer cells, coincident with increased DNA damage. Seeking natural compounds as BRD9 antagonists, molecular docking in silico identified several polyphenols such as Epigallocatechin-3-gallate (EGCG), Equol, Quercetin, and Aspalathin, with favorable binding energies, supported by BROMOscan® (DiscoverX) and isothermal titration calorimetry experiments. Polyphenols mimicked BRD9 knockdown and iBRD9 treatment in reducing colon cancer cell viability, inhibiting colony formation, and enhancing DNA damage and apoptosis. Normal colonic epithelial cells were unaffected, signifying cancer-specific effects. These findings suggest that natural polyphenols recognize and target BRD9 for inhibition, and might serve as useful lead compounds for bromodomain therapeutics in the clinical setting.


Asunto(s)
Antineoplásicos , Neoplasias del Colon , Humanos , Polifenoles/farmacología , Simulación del Acoplamiento Molecular , ARN Interferente Pequeño , Equol , Quercetina , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Apoptosis , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Daño del ADN
3.
Cells ; 11(3)2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35159382

RESUMEN

There is growing interest in the crosstalk between the gut microbiome, host metabolomic features, and disease pathogenesis. The current investigation compared long-term (26 week) and acute (3 day) dietary spinach intake in a genetic model of colorectal cancer. Metabolomic analyses in the polyposis in rat colon (Pirc) model and in wild-type animals corroborated key contributions to anticancer outcomes by spinach-derived linoleate bioactives and a butanoate metabolite linked to increased α-diversity of the gut microbiome. Combining linoleate and butanoate metabolites in human colon cancer cells revealed enhanced apoptosis and reduced cell viability, paralleling the apoptosis induction in colon tumors from rats given long-term spinach treatment. Mechanistic studies in cell-based assays and in vivo implicated the linoleate and butanoate metabolites in targeting histone deacetylase (HDAC) activity and the interferon-γ (IFN-γ) signaling axis. Clinical translation of these findings to at-risk patients might provide valuable quality-of-life benefits by delaying surgical interventions and drug therapies with adverse side effects.


Asunto(s)
Ácido Butírico , Neoplasias del Colon , Dieta , Ácido Linoleico , Spinacia oleracea , Animales , Neoplasias del Colon/patología , Humanos , Interferón gamma/uso terapéutico , Metabolómica , Ratas
4.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R678-R686, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30892915

RESUMEN

Obesity is the only known modifiable risk factor for multiple myeloma (MM), an incurable cancer of bone marrow plasma cells. The mechanism linking the two is unknown. Obesity is associated with an increased risk of sleep apnea, which results in chronic intermittent hypoxia (CIH), and drives solid tumor aggressiveness. Given the link between CIH and solid tumor progression, we tested the hypothesis that CIH drives the proliferation of MM cells in culture and their engraftment and progression in vivo. Malignant mouse 5TGM1 cells were cultured in CIH, static hypoxia, or normoxia as a control in custom, gas-permeable plates. Typically MM-resistant C57BL/6J mice were exposed to 10 h/day CIH (AHI = 12/h), static hypoxia, or normoxia for 7 days, followed by injection with 5TGM1 cells and an additional 28 days of exposure. CIH and static hypoxia slowed the growth of 5TGM1 cells in culture. CIH-exposed mice developed significantly more MM than controls (67 vs. 12%, P = 0.005), evidenced by hindlimb paralysis, gammopathy, bone lesions, and bone tumor formation. Static hypoxia was not a significant driver of MM progression and did not reduce survival (P = 0.117). Interestingly, 5TGM1 cells preferentially engrafted in the bone marrow and promoted terminal disease in CIH mice, despite a lower tumor burden, compared with the positive controls. These first experiments in the context of hematological cancer demonstrate that CIH promotes MM through mechanisms distinct from solid tumors and that sleep apnea may be a targetable risk factor in patients with or at risk for blood cancer.


Asunto(s)
Proliferación Celular , Hipoxia/complicaciones , Mieloma Múltiple/patología , Animales , Línea Celular Tumoral , Enfermedad Crónica , Progresión de la Enfermedad , Femenino , Hipoxia/metabolismo , Ratones Endogámicos C57BL , Mieloma Múltiple/complicaciones , Mieloma Múltiple/metabolismo , Factores de Tiempo , Carga Tumoral , Hipoxia Tumoral , Microambiente Tumoral
5.
FASEB Bioadv ; 1(7): 404-414, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32095781

RESUMEN

The orphan small nucleolar RNA (snoRNA) ACA11 is overexpressed as a result of the t(4;14) chromosomal translocation in multiple myeloma (MM), increases reactive oxygen species, and drives cell proliferation. Like other snoRNAs, ACA11 is predominantly localized to a sub-nuclear organelle, the nucleolus. We hypothesized that increased ACA11 expression would increase ribosome biogenesis and protein synthesis. We found that ACA11 overexpression in MM cells increased nucleolar area and number as well as silver-binding nucleolar organizing regions (AgNORs). Supporting these data, samples from t(4;14)-positive patients had higher AgNORs scores than t(4;14)-negative samples. ACA11 also upregulated ribosome production, pre-47S rRNA synthesis, and protein synthesis in a ROS-dependent manner. Lastly, ACA11 overexpression enhanced the response to proteasome inhibitor in MM cells, while no effect was found in response to high doses of melphalan. Together, these data demonstrate that ACA11 stimulates ribosome biogenesis and influences responses to chemotherapy. ACA11 may be a useful target to individualize the treatment for t(4;14)-positive myeloma patients.

6.
Oncoimmunology ; 5(7): e1196299, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27622050

RESUMEN

The potential of a tumor cell to metastasize profoundly depends on its microenvironment, or "niche" interactions with local components. Tumor-associated-macrophages (TAMs) are the most abundant subpopulation of tumor stroma and represent a key component of tumor microenvironment. The dynamic interaction of cancer cells with neighboring TAMs actively drive cancer progression and metastatic transformation through intercellular signaling networks that need better elucidation. Thus, current study was planned for discerning paracrine communication networks operational between TAMs, and breast cancer cells with special reference to cancer cell invasion and dissemination to distant sites. Here, we report role of MIP-1ß in enhancing invasive potential of metastatic breast cancer MDA-MB-231 and MDA-MB-468 cells. In addition, the poorly metastatic MCF-7 cells were also rendered invasive by MIP-1ß. The MIP-1ß-driven cancer cell invasion was dependent on upregulated expression levels of MYO3A gene, which encodes an unconventional myosin super-family protein harboring a kinase domain. Ex ovo study employing Chick-embryo-model and in vivo Syngenic 4T1/BALB/c mice-model further corroborated aforementioned in vitro findings, thereby substantiating their physiological relevance. Concordantly, human breast cancer specimen exhibited significant association between mRNA expression levels of MIP-1ß and MYO3A. Both, MIP-1ß and MYO3A exhibited positive correlation with MMP9, an established molecular determinant of cancer cell invasion. Higher expression of these genes correlated with poor survival of breast cancer patients. Collectively, these results point toward so far undisclosed MIP-1ß/MYO3A axis being operational during metastasis, wherein macrophage-derived MIP-1ß potentiated cancer cell invasion and metastasis via up regulation of MYO3A gene within cancer cells. Our study exposes opportunities for devising potential anti-metastatic strategies for efficient clinical management of breast cancer.

7.
Oncotarget ; 5(14): 5350-68, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25051364

RESUMEN

TAMs, a unique and distinct M2-skewed myeloid population of tumor stroma, exhibiting pro-tumor functions is fast emerging as a potential target for anti-cancer immunotherapy. Macrophage-recruitment and M2-polarization represent key TAMs-related phenomenon that are amenable to therapeutic intervention. However successful translation of these approaches into effective therapeutic regimen requires better characterization of tumor-microenvironment derived signals that regulate macrophage recruitment and their polarization. Owing to hypoxic milieu being a persistent feature of tumor-microenvironment and a major contributor to malignancy and treatment resistance, the current study was planned with an aim to decipher tumor cell responses to hypoxia vis-a-vis macrophage homing and phenotype switching. Here, we show that hypoxia-primed cancer cells chemoattract and polarize macrophages to pro-angiogenic M2-polarized subtype via Eotaxin and Oncostatin M. Concordantly, hypoxic regions of human breast-cancer specimen exhibited elevated Eotaxin and Oncostatin M levels with concurrently elevated M2-macrophage content. Blockade of Eotaxin/Oncostatin M not only prevented hypoxic breast-cancer cells from recruiting and polarizing macrophages towards an M2-polarized phenotype and retarded tumor progression in 4T1/BALB/c-syngenic-mice-model of breast-cancer but also enhanced the efficacy of anti-angiogenic Bevacizumab. The findings established these two cytokines as novel targets for devising effective anticancer therapy particularly for tumors that are refractory or develop resistance to anti-angiogenic therapeutics.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Quimiocina CCL11/metabolismo , Macrófagos/patología , Oncostatina M/metabolismo , Animales , Neoplasias de la Mama/patología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fenotipo , Distribución Aleatoria , Microambiente Tumoral
8.
PLoS One ; 8(6): e67586, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23799149

RESUMEN

FasL mediated preferential apoptosis of bystander CTLs while protection of infected CD4(+)T cells remains one of the hallmarks of immune evasion during HIV infection. The property of infected host cells to evade cell-autonomous apoptosis emanates from ability of HIV-1Nef-protein to physically interact with ASK-1 and thereby inhibit its enzymatic activity. The specific domains of HIV-1Nef through which it may interact with ASK1 and thereby impair the ASK1 activity remain unidentified so far and represent a major challenge towards developing clear understanding about the dynamics of this interaction. Using mammalian two hybrid screen in association with site directed mutagenesis and competitive inhibitor peptides, we identified constituent minimal essential domain (152 DEVGEANN 159) through which HIV-1Nef interacts with ASK1 and inhibits its function. Furthermore our study also unravels a novel alternate mechanism underlying HIV-1 Nef mediated ASK1 functional modulation, wherein by potentiating the inhibitory ser(967) phosphorylation of ASK1, HIV-1Nef negatively modulated ASK1 function.


Asunto(s)
VIH-1/fisiología , MAP Quinasa Quinasa Quinasa 5/química , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/química , Secuencia de Aminoácidos , Apoptosis , Sitios de Unión , Unión Competitiva , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , MAP Quinasa Quinasa Quinasa 5/metabolismo , Fragmentos de Péptidos/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Técnicas del Sistema de Dos Híbridos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
9.
Free Radic Biol Med ; 53(10): 1929-41, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23000059

RESUMEN

The estrogen receptor (ER) plays a cardinal role in estrogen-responsive breast carcinogenesis. It is, however, unclear as to how estrogen-ER interaction potentiates breast cancer progression. Compelling evidence supports estrogen-induced redox alterations, such as augmented reactive oxygen species (ROS) levels, as having a crucial role in breast carcinogenesis. Despite ER being a biological mediator of the majority of estrogen-induced cellular responses; its role in estrogen-induced tissue-specific ROS generation remains largely debatable. We examined a panel of human breast cancer specimens and found that ER-positive breast cancer specimens exhibited a higher incidence of augmented O(2)(•-) levels compared to matched normal tissue. ROS are known to function as signal transducers and ROS-mediated signaling remains a key complementary mechanism that drives carcinogenesis by activating redox-sensitive oncogenic pathways. Additional studies revealed that augmented O(2)(•-) levels in breast cancer specimens coincided with mammalian target of rapamycin complex 2 (mTORC2) hyperactivation. Detailed investigations using in vitro experiments established that 17ß-estradiol (E2)-stimulated breast cancer cells exhibited transiently upregulated O(2)(•-) levels, with the presence of ER being a crucial determinant for the phenomenon to take place. Gene expression, ER transactivation, and confocal studies revealed that the E2-induced transient O(2)(•-) upregulation was effected by ER through a nongenomic pathway possibly involving mitochondria. Furthermore, E2 treatment activated mTORC2 in breast cancer cells in a characteristically ER-dependent manner. Interestingly, altering O(2)(•-) anion levels through chemical/genetic methods caused significant modulation of the mTORC2 signaling cascade. Taken together, our findings unravel a novel nongenomic pathway unique to estrogen-responsive breast cancer cells wherein, upon stimulation by E2, ER may regulate mTORC2 activity in a redox-dependent manner by transiently modulating O(2)(•-) levels particularly within mitochondria. The findings suggest that therapies aimed at counteracting these redox alterations and/or resultant signaling cascades may complement conventional treatments for estrogen-responsive breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Complejos Multiproteicos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/metabolismo , Superóxidos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Estradiol/farmacología , Femenino , Humanos , Células MCF-7 , Diana Mecanicista del Complejo 2 de la Rapamicina , Mitocondrias/metabolismo , Oxidación-Reducción , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/fisiología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba
10.
Toxicol Mech Methods ; 20(5): 242-51, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20433247

RESUMEN

Isoniazid (INH) is a first-line antibiotic used in the treatment of infections caused by Mycobacterium tuberculosis. However it has a serious limitation of being hepatotoxic. Delineating the mechanism underlying INH-induced hepatotoxicity may be beneficial in devising ways to counteract its toxic manifestations. Studies in human hepatoma HepG2 cells have indicated that INH exposure causes induction of apoptosis. This study was aimed at identifying the key components/pathways of the INH-induced apoptotic pathway using HepG2 cells. HepG2 cells were exposed to increasing concentrations of INH (6.5, 13, 26, and 52 mM). Hydrogen peroxide (0.3 mM) served as positive control. After incubating for specific time intervals cells were harvested and evidences of cytotoxicity, oxidative stress, and apoptosis were sought. The findings indicated that INH exposure causes increased ROS generation along with alteration in levels of enzymatic antioxidants such as Superoxide dismutase, Catalase, and Glucose-6-Phosphate dehydrogenase. Altered Bcl-2/Bax content, cytochrome-c translocation, caspase activation, and DNA fragmentation emphasized involvement of apoptosis.


Asunto(s)
Antituberculosos/toxicidad , Apoptosis , Isoniazida/toxicidad , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Catalasa/metabolismo , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Glucosafosfato Deshidrogenasa/metabolismo , Células Hep G2 , Humanos , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...