Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioorg Med Chem ; 81: 117193, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36796126

RESUMEN

Oral squamous cell carcinoma (OSCC) is the most common malignant epithelial neoplasm, affects the mouth and throat, and accounts for 90 % of oral cancers. Considering the associated morbidity with neck dissections and the limitation of existing therapeutic agents, the discovery and development of new anticancer drugs/drug candidates for oral cancer treatment are of the utmost need. In this context, reported here is the identification of fluorinated 2­styryl 4(3H)-quinazolinone as a promising hit for oral cancer. Preliminary studies indicate that the compound blocks the transition of G1 to S phase, thereby leading to arrest in the G1/S phase. Subsequent RNA-seq analysis revealed that the compound induces the activation of molecular pathways involved in apoptosis (such as TNF signalling through NF-κB, p53 pathways) and cell differentiation and suppresses the pathways of cellular growth and development (such as KRAS signaling) in CAL-27 cancer cells. It is noted that identified hit complies with a favorable range of ADME properties as per the computational analysis.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de la Boca , Humanos , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/patología , Transducción de Señal
2.
J Biol Chem ; 293(22): 8330-8341, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29636415

RESUMEN

Diacylglycerol (DAG) is a key lipid second messenger downstream of cellular receptors that binds to the C1 domain in many regulatory proteins. Protein kinase C (PKC) isoforms constitute the most prominent family of signaling proteins with DAG-responsive C1 domains, but six other families of proteins, including the chimaerins, Ras-guanyl nucleotide-releasing proteins (RasGRPs), and Munc13 isoforms, also play important roles. Their significant involvement in cancer, immunology, and neurobiology has driven intense interest in the C1 domain as a therapeutic target. As with other classes of targets, however, a key issue is the establishment of selectivity. Here, using [3H]phorbol 12,13-dibutyrate ([3H]PDBu) competition binding assays, we found that a synthetic DAG-lactone, AJH-836, preferentially binds to the novel PKC isoforms PKCδ and PKCϵ relative to classical PKCα and PKCßII. Assessment of intracellular translocation, a hallmark for PKC activation, revealed that AJH-836 treatment stimulated a striking preferential redistribution of PKCϵ to the plasma membrane relative to PKCα. Moreover, unlike with the prototypical phorbol ester phorbol 12-myristate 13-acetate (PMA), prolonged exposure of cells to AJH-836 selectively down-regulated PKCδ and PKCϵ without affecting PKCα expression levels. Biologically, AJH-836 induced major changes in cytoskeletal reorganization in lung cancer cells, as determined by the formation of membrane ruffles, via activation of novel PKCs. We conclude that AJH-836 represents a C1 domain ligand with PKC-activating properties distinct from those of natural DAGs and phorbol esters. Our study supports the feasibility of generating selective C1 domain ligands that promote novel biological response patterns.


Asunto(s)
Diglicéridos/química , Lactonas/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Células A549 , Unión Competitiva , Células HeLa , Humanos , Ligandos , Unión Proteica , Transporte de Proteínas , Especificidad por Sustrato
3.
J Biol Chem ; 289(28): 19823-38, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24825907

RESUMEN

Overexpression of PKCϵ, a kinase associated with tumor aggressiveness and widely implicated in malignant transformation and metastasis, is a hallmark of multiple cancers, including mammary, prostate, and lung cancer. To characterize the mechanisms that control PKCϵ expression and its up-regulation in cancer, we cloned an ∼ 1.6-kb promoter segment of the human PKCϵ gene (PRKCE) that displays elevated transcriptional activity in cancer cells. A comprehensive deletional analysis established two regions rich in Sp1 and STAT1 sites located between -777 and -105 bp (region A) and -921 and -796 bp (region B), respectively, as responsible for the high transcriptional activity observed in cancer cells. A more detailed mutagenesis analysis followed by EMSA and ChIP identified Sp1 sites in positions -668/-659 and -269/-247 as well as STAT1 sites in positions -880/-869 and -793/-782 as the elements responsible for elevated promoter activity in breast cancer cells relative to normal mammary epithelial cells. RNAi silencing of Sp1 and STAT1 in breast cancer cells reduced PKCϵ mRNA and protein expression, as well as PRKCE promoter activity. Moreover, a strong correlation was found between PKCϵ and phospho-Ser-727 (active) STAT1 levels in breast cancer cells. Our results may have significant implications for the development of approaches to target PKCϵ and its effectors in cancer therapeutics.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Proteína Quinasa C-epsilon/biosíntesis , Elementos de Respuesta , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción Sp1/metabolismo , Transcripción Genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Proteínas de Neoplasias/genética , Proteína Quinasa C-epsilon/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción Sp1/genética
4.
J Biol Chem ; 287(44): 37570-82, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22955280

RESUMEN

Protein kinase C ε (PKCε) has emerged as an oncogenic kinase and plays important roles in cell survival, mitogenesis and invasion. PKCε is up-regulated in most epithelial cancers, including prostate, breast, and lung cancer. Here we report that PKCε is an essential mediator of NF-κB activation in prostate cancer cells. A strong correlation exists between PKCε overexpression and NF-κB activation status in prostate cancer cells. Moreover, transgenic overexpression of PKCε in the mouse prostate causes preneoplastic lesions that display significant NF-κB hyperactivation. PKCε RNAi depletion or inhibition in prostate cancer cells diminishes NF-κB translocation to the nucleus with subsequent impairment of both activation of NF-κB transcription and induction of NF-κB responsive genes in response to the proinflammatory cytokine tumor necrosis factor α (TNFα). On the other hand, PKCε overexpression in normal prostate cells enhances activation of the NF-κB pathway. A mechanistic analysis revealed that TNFα activates PKCε via a C1 domain/diacylglycerol-dependent mechanism that involves phosphatidylcholine-phospholipase C. Moreover, PKCε facilitates the assembly of the TNF receptor-I signaling complex to trigger NF-κB activation. Our studies identified a molecular link between PKCε and NF-κB that controls key responses implicated in prostate cancer progression.


Asunto(s)
Neoplasias de la Próstata/enzimología , Proteína Quinasa C-epsilon/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Proteínas I-kappa B/metabolismo , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Masculino , Ratones , Ratones Transgénicos , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Neoplasias de la Próstata/metabolismo , Unión Proteica , Proteína Quinasa C-epsilon/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Fosfolipasas de Tipo C/metabolismo
5.
Cancer Lett ; 557: 216079, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36736532

RESUMEN

Cancer is a pervasive, constantly evolving, and significant public health concern. The number of new cancer cases has risen dramatically in the last decades, making it one of the top causes of poor health and mortality worldwide. Although various treatment strategies, including surgery, radiation, and pharmaceutical therapies, have evolved into more sophisticated, precise methods, there is not much improvement in the cancer-related death toll. Consequently, natural product-based therapeutic discoveries have recently been considered an alternative approach. According to an estimate, one-third of the top twenty medications in today's market have a natural plant-product-based origin. Accordingly, primary prevention is an essential component of worldwide cancer control. This review provides an overview of the mechanisms of action of bioactive ingredients in natural dietary products that may contribute to the prevention and management of multiple malignancies.


Asunto(s)
Productos Biológicos , Neoplasias , Humanos , Pronóstico , Productos Biológicos/uso terapéutico , Neoplasias/tratamiento farmacológico
6.
Cancers (Basel) ; 15(11)2023 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-37296923

RESUMEN

Animal models have been utilized for decades to investigate the causes of human diseases and provide platforms for testing novel therapies. Indeed, breakthrough advances in genetically engineered mouse (GEM) models and xenograft transplantation technologies have dramatically benefited in elucidating the mechanisms underlying the pathogenesis of multiple diseases, including cancer. The currently available GEM models have been employed to assess specific genetic changes that underlay many features of carcinogenesis, including variations in tumor cell proliferation, apoptosis, invasion, metastasis, angiogenesis, and drug resistance. In addition, mice models render it easier to locate tumor biomarkers for the recognition, prognosis, and surveillance of cancer progression and recurrence. Furthermore, the patient-derived xenograft (PDX) model, which involves the direct surgical transfer of fresh human tumor samples to immunodeficient mice, has contributed significantly to advancing the field of drug discovery and therapeutics. Here, we provide a synopsis of mouse and zebrafish models used in cancer research as well as an interdisciplinary 'Team Medicine' approach that has not only accelerated our understanding of varied aspects of carcinogenesis but has also been instrumental in developing novel therapeutic strategies.

7.
Cancers (Basel) ; 14(23)2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36497402

RESUMEN

N6-methyladenosine (m6A) is the most prevalent post-transcriptional RNA modification regulating cancer self-renewal. However, despite its functional importance and prognostic implication in tumorigenesis, the relevance of FTO, an m6A eraser, in pancreatic cancer (PC) remains elusive. Here, we establish the oncogenic role played by FTO overexpression in PC. FTO is upregulated in PC cells compared to normal human pancreatic ductal epithelial (HPDE) cells. Both RNAi depletion and CS1-mediated pharmacological inhibition of FTO caused a diminution of PC cell proliferation via cell cycle arrest in the G1 phase and p21cip1 and p27kip1 induction. While HPDE cells remain insensitive to CS1 treatment, FTO overexpression confers enhancements in growth, motility, and EMT transition, thereby inculcating tumorigenic properties in HPDE cells. Notably, shRNA-mediated FTO depletion in PC cells impairs their mobility and invasiveness, leading to EMT reversal. Mechanistically, this was associated with impaired tumorsphere formation and reduced expression of CSCs markers. Furthermore, FTO depletion in PC cells weakened their tumor-forming capabilities in nude mice; those tumors had increased apoptosis, decreased proliferation markers, and MET conversion. Collectively, our study demonstrates the functional importance of FTO in PC and the maintenance of CSCs via EMT regulation. Thus, FTO may represent an attractive therapeutic target for PC.

8.
Front Oncol ; 12: 855860, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35600365

RESUMEN

Background: Naturally occurring dietary botanicals offer time-tested safety and anti-cancer efficacy, and a combination of certain compounds has shown to overcome the elusive chemotherapeutic resistance, which is of great significance for improving the mortality of patients with colorectal cancer (CRC). Accordingly, herein, we hypothesized that berberine (BBR) and oligomeric proanthocyanidins (OPCs) might regulate synergistically multiple oncogenic pathways to exert a superior anti-cancer activity in CRC. Methods: We performed a series of cell culture studies, followed by their interrogation in patient-derived organoids to evaluate the synergistic effect of BBR and OPCs against CRC. In addition, by performing whole genome transcriptomic profiling we identified the key targeted genes and pathways regulated by the combined treatment. Results: We first demonstrated that OPCs facilitated enhanced cellular uptake of BBR in CRC cells by measuring the fluorescent signal of BBR in cells treated individually or their combination. The synergism between BBR and OPCs were investigated in terms of their anti-tumorigenic effect on cell viability, clonogenicity, migration, and invasion. Furthermore, the combination treatment potentiated the cellular apoptosis in an Annexin V binding assay. Transcriptomic profiling identified oncogene MYB in PI3K-AKT signaling pathway might be critically involved in the anti-tumorigenic properties of the combined treatment. Finally, we successfully validated these findings in patient-derived CRC tumor organoids. Conclusions: Collectively, we for the first time demonstrate that a combined treatment of BBR and OPCs synergistically promote the anti-tumorigenic properties in CRC possibly through the regulation of cellular apoptosis and oncogene MYB in the PI3K-Akt signaling pathway.

10.
Biochim Biophys Acta Rev Cancer ; 1877(5): 188803, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36150564

RESUMEN

Previously, we showed that knockout mice homozygous for deficiency of the mercapturic acid pathway (MAP) transporter protein, RLIP (RLIP-/-), are resistant to chemical carcinogenesis, inflammation, and metabolic syndrome (MetS). We also found that RLIP-/- mice are highly resistant to obesity caused by a high-fat diet (HFD). Interestingly, these studies showed that kinase, cytokine, and adipokine signaling that are characteristics of obesity were blocked despite the presence of increased oxidative stress in RLIP-/- mice. The deficiencies in obesity-inducing kinase, cytokine, and adipokine signaling were attributable to a lack of clathrin-dependent endocytosis (CDE), a process that is severely deficient in RLIP-/- mice. Because CDE is also necessary for carcinogenic signaling through EGF, WNT, TGFß and other cancer-specific peptide hormones, and because RLIP-/- mice are cancer-resistant, we reasoned that depletion of RLIP by an antisense approach should cause cancer regression in human cancer xenografts. This prediction has been confirmed in studies of xenografts from lung, kidney, prostate, breast, and pancreatic cancers and melanoma. Because these results suggested an essential role for RLIP in carcinogenesis, and because our studies have also revealed a direct interaction between p53 and RLIP, we reasoned that if RLIP played a central role in carcinogenesis, that development of lymphoma in p53-/- mice, which normally occurs by the time these mice are 6 months old, could be delayed or prevented by depleting RLIP. Recent studies described herein have confirmed this hypothesis, showing complete suppression of lymphomagenesis in p53-/- mice treated with anti-RLIP antisense until the age of 8 months. All control mice developed lymphoma in the thymus or testis as expected. These findings lead to a novel paradigm predicting that under conditions of increased oxidative stress, the consequent increased flux of metabolites in the MAP causes a proportional increase in the rate of CDE. Because CDE inhibits insulin and TNF signaling but promotes EGF, TGFß, and Wnt signaling, our model predicts that chronic stress-induced increases in RLIP (and consequently CDE) will induce insulin-resistance and enhance predisposition to cancer. Alternatively, generalized depletion of RLIP would antagonize the growth of malignant cells, and concomitantly exert therapeutic insulin-sensitizing effects. Therefore, this review focuses on how targeted depletion or inhibition of RLIP could provide a novel target for treating both obesity and cancer.


Asunto(s)
Insulinas , Neoplasias , Hormonas Peptídicas , Acetilcisteína/metabolismo , Adipoquinas/metabolismo , Animales , Carcinogénesis/genética , Carcinógenos , Proteínas Portadoras/metabolismo , Clatrina/metabolismo , Citocinas/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Lactante , Insulinas/metabolismo , Masculino , Ratones , Obesidad/genética , Estrés Oxidativo , Hormonas Peptídicas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
11.
ISA Trans ; 100: 251-263, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31955950

RESUMEN

The conventional maximum power point tracking (MPPT) algorithm shows best performance under uniform insolation but when photovoltaic (PV) array is partially irradiated, the Power vs Voltage (P-V) plot consists of multiple local maxima power point (LMPP) and one global maxima power point (GMPP). The conventional MPPT algorithm may track local peak and fluctuate around it, resulting in lower power tracking. To eradicate this drawback of conventional algorithm, the solar PV system requires the synthesis of modified controller which is able to discriminate between local and global peak point. Along with implementing modified MPPT controller, to minimise the adverse effect of partial shading on PV system, different PV array arrangements like series-parallel (SP), honey comb (HC), total cross tied (TCT) etc. may be used. Author(s) in the present study, has proposed asymmetrical interval type-2 fuzzy logic control (IT-2 AFLC) based MPP algorithm for tracking global peak in partial shading condition (PSC) with different PV array arrangements. The presented algorithm has been compared with other approaches viz. perturb & observe (P&O) and type-1(T-1) FLC for GMPP tracking, fill factor, shading losses, mismatch loss and efficiency to establish its superiority. For evaluating the efficiency of different algorithms, the EN50530 MPPT efficiency test has been performed under dynamic condition. The proposed algorithm has been developed under MATLAB/Simulink environment.

12.
Oncotarget ; 11(13): 1157-1171, 2020 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-32284792

RESUMEN

Cisplatin is used as chemotherapeutic drug for oral squamous cell carcinoma (OSCC). However, OSCC cells develop resistance following long-term cisplatin exposure. Resistance against cisplatin chemo-therapy is accredited to the process of epithelial-to-mesenchymal transition, which in-turn has been linked to tumor-recurrence. miRNA deregulation, a common event in cancer, plays contributory role in chemo-resistance. Exosomes acts as the natural cargo for miRNA and facilitates inter-cell communication in the tumor micro-environment. Hence, exosomal-mediated miRNA transference may play essential role in drug resistance and serve as a target for cancer-therapy. miR-155 upregulation in OSCC has been described, however, its relevance in the observed chemo-resistance is unclear and also, if exosomes have any role in miR-155 regulation remain elusive. In the present study, we document for the first time the critical role of exosomes in mediating increments in miR-155 expression in OSCC cells that have acquired cisplatin resistance (cisRes cells). Importantly, exosomal transfer from cisRes to the cisplatin sensitive (cisSens) cells was found to confer significant miR-155 induction in the recipient cisSens cells. Restoration of miR-155 expression in cisSens cells following miR-155 mimics treatment led to epithelial to mesenchymal transition, enhancements in their migratory potential as well as acquisition of resistant phenotype. Notably, similar augmentations in the migratory and chemo-resistant traits were seen upon delivery of exosomes from cisRes to the recipient cisSens cells. Overall, our findings establish the significance of exosomal-mediated miR-155 shuttling in the cisplatin-chemoresistance, commonly observed in OSCC cells, thereby providing rationale for targeting miR-155 signalling for oral cancer therapy.

13.
Oncotarget ; 11(20): 1832-1845, 2020 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-32499869

RESUMEN

Exosomes facilitate cross-talk amongst tumor cells, and thus also possess the potential to influence tumor-microenvironment and chemo-resistance. miRNAs, the important constituent of exosomes, are often dysregulated in cancer. They have been shown to play an essential role in tumor progression, metastasis, invasion, and resistance developed against different therapies. Acquisition of cisplatin-chemoresistance remains a major hurdle in the effective treatment of oral squamous cell carcinoma (OSCC). In this study, we demonstrate the importance of exosome-mediated miR-30a transfer in conferring cisplatin sensitivity in the otherwise resistant OSCC cells. Notably, miR-30a was found to be significantly reduced in exosomes isolated from the serum of OSCC patients, especially those having disease-recurrence, post cisplatin treatment. In conjunction with the findings in clinical samples, decreased miR-30a expression was observed in vitro in the cisplatin-resistant cultured OSCC cells compared to the cisplatin-sensitive cells. Besides, we identified Beclin1, an autophagy-related marker, as a target of miR-30a and found it to be overexpressed in cisplatin-resistant OSCC cells, thus indicating at its possible negative-regulation by miR30a. Exosomes from the cisplatin-resistant cells that have been transfected with miR-30a mimics, when delivered to the naïve cisplatin-resistant cells, caused not only the significant enhancements in miR-30a expression but also a concomitant decrease in Beclin1 and Bcl2 expression (autophagic and anti-apoptotic marker). More importantly, this together resulted in the sensitization of cisplatin-resistant cells. Thus, our study highlighted the role of exosomal-mediated miR-30a transfer in regaining sensitivity of the cisplatin-resistant OSCC cells via Beclin1 and Bcl2 regulation and hence suggests at its potential therapeutic role.

14.
Cancer Res ; 80(23): 5166-5173, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32994205

RESUMEN

Non-small cell lung cancer (NSCLC) is the most frequent subtype of lung cancer and remains a highly lethal malignancy and one of the leading causes of cancer-related deaths worldwide. Mutant KRAS is the prevailing oncogenic driver of lung adenocarcinoma, the most common histologic form of NSCLC. In this study, we examined the role of PKCϵ, an oncogenic kinase highly expressed in NSCLC and other cancers, in KRAS-driven tumorigenesis. Database analysis revealed an association between PKCϵ expression and poor outcome in patients with lung adenocarcinoma specifically harboring KRAS mutations. A PKCϵ-deficient, conditionally activatable allele of oncogenic Kras (LSL-KrasG12D ;PKCϵ-/- mice) demonstrated the requirement of PKCϵ for Kras-driven lung tumorigenesis in vivo, which was consistent with impaired transformed growth reported in PKCϵ-deficient KRAS-dependent NSCLC cells. Moreover, PKCϵ-knockout mice were found to be less susceptible to lung tumorigenesis induced by benzo[a]pyrene, a carcinogen that induces mutations in Kras. Mechanistic analysis using RNA sequencing revealed little overlap for PKCϵ and KRAS in the control of genes and biological pathways relevant in NSCLC, suggesting that a permissive role of PKCϵ in KRAS-driven lung tumorigenesis may involve nonredundant mechanisms. Our results thus, highlight the relevance and potential of targeting PKCϵ for lung cancer therapeutics. SIGNIFICANCE: These findings demonstrate that KRAS-mediated tumorigenesis requires PKCϵ expression and highlight the potential for developing PKCϵ-targeted therapies for oncogenic RAS-driven malignancies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Proteína Quinasa C-epsilon/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/mortalidad , Adenocarcinoma del Pulmón/patología , Animales , Benzo(a)pireno/toxicidad , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Ratones Noqueados , Ratones Transgénicos , Mutación , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Proteína Quinasa C-epsilon/genética
15.
Carcinogenesis ; 29(5): 1022-32, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18321868

RESUMEN

Curcumin has been shown to possess anti-initiating and anti-promoting activity in experimental systems. However, the mechanisms of its actions are not fully elucidated in vivo. In the present study, mechanisms of curcumin-mediated anti-initiation were investigated in mice employing benzo[a]pyrene (B[a]P) as a model carcinogen. Dietary pretreatment of mice with chemopreventive doses of curcumin showed significant inhibition of B[a]P-induced enzyme activity, protein and messenger RNA (mRNA) levels of cytochrome P450 1A1/1A2 in liver and lungs. Although curcumin alone did not alter the basal levels of aryl hydrocarbon receptor (AhR), it significantly decreased the B[a]P-induced AhR protein levels, its phosphorylation, nuclear translocation and subsequent binding to DNA, thereby decreasing the transactivation of CYP1A. Dietary curcumin led to increase in NF-E2-related factor-2 (Nrf2) protein levels and enhanced its nuclear translocation in liver and lungs of mice as compared with controls. Additionally, increased binding of Nrf2 to antioxidant response element occurred in nuclear extracts from liver and lungs of mice pretreated with dietary curcumin. Induction of activity, protein and mRNA levels of glutathione S-transferase, its isoforms and NAD(P)H:quinone oxidoreductase-1 by dietary curcumin in mice paralleled the curcumin-mediated activation of Nrf2, leading to increased detoxification of B[a]P. In agreement with the observed curcumin-mediated decrease in B[a]P-induced phase I enzyme and concomitant induction of phase II enzymes, pretreatment with dietary curcumin resulted in significant reduction of B[a]P-induced DNA adduct, oxidative damage and inflammation. To conclude, curcumin exhibits anti-initiating effects via modulating the transcriptional regulators of phase I and phase II enzymes in mice.


Asunto(s)
Benzo(a)pireno/toxicidad , Curcumina/farmacología , Transcripción Genética/efectos de los fármacos , Alimentación Animal , Animales , Carcinógenos/toxicidad , Curcumina/administración & dosificación , Aductos de ADN/metabolismo , Dieta , Glutatión Transferasa/efectos de los fármacos , Glutatión Transferasa/metabolismo , Pulmón/enzimología , Ratones , Microsomas/efectos de los fármacos , Microsomas/enzimología , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Carcinogenesis ; 29(6): 1249-57, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18477648

RESUMEN

Curcumin has been shown to inhibit 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced tumour promotion and some of the TPA-responsive markers in mouse skin. However, its mechanism of action is not fully elucidated. The present study focuses on understanding the role of protein kinase C (PKC), the major cellular receptor of TPA, in mediating TPA-induced biological responses in mouse skin and subsequently, elucidating the effects of curcumin on PKC and its downstream target molecules. As compared with controls, single topical application of TPA (5 nmol) to skin increased the translocation of PKC from cytosolic to particulate fraction, determined in terms of activity and protein levels. Ro-31- 8220 (PKC inhibitor, 1 nmol) when applied topically, alone or prior to TPA, inhibited PKC activity in both the compartments but did not affect the TPA-induced protein translocation. In contrast, though curcumin (10 mumol) alone did not alter the basal activity/levels, its pre-treatment decreased the TPA-induced translocation of PKC isozymes (alpha, beta, gamma, epsilon, eta), resulting in appropriate alterations in activity. Despite differences in modes of action of Ro-31-8220 (activity inhibition) and curcumin (decreasing translocation) in modulating PKC, their pre-treatment blunted the TPA-induced levels of mitogen-activated protein kinases and transcription factors (c-jun, c-fos and nuclear factor-kappa B) and downstream target proteins associated with cell proliferation (cyclin D1 and ornithine decarboxylase), cell death (Bax and Bcl2), inflammation (cyclooxygenase-2 and prostaglandin E2) and oxidative stress (8-hydroxy-2'-deoxyguanosine) in skin. These results demonstrate the crucial role of PKC in TPA-mediated cellular responses in skin and that curcumin modulates transmembrane signal transduction via PKC to affect TPA-induced biochemical and molecular alterations in mouse skin.


Asunto(s)
Antineoplásicos/farmacología , Curcumina/farmacología , Proteína Quinasa C/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos , Acetato de Tetradecanoilforbol/toxicidad , Animales , Ensayo de Cambio de Movilidad Electroforética , Inhibidores Enzimáticos/farmacología , Femenino , Immunoblotting , Isoenzimas/efectos de los fármacos , Isoenzimas/metabolismo , Ratones , Ratones Desnudos , Proteína Quinasa C/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/fisiología , Piel/metabolismo
17.
Toxicol Appl Pharmacol ; 232(3): 428-39, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18687351

RESUMEN

The chemopreventive efficacy of turmeric has been established in experimental systems. However, its mechanism(s) of action are not fully elucidated in vivo. The present study investigates the mechanism of turmeric-mediated chemoprevention in 7,12-dimethylbenz(a)anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinogenesis at 2, 4, 6, 10 and 12 weeks. Dietary turmeric (1%) led to decrease in DMBA-induced tumor burden and multiplicity, and enhanced the latency period in parallel, to its modulatory effects on oncogene products and various cellular responses during HBP tumorigenesis. DMBA-induced expression of ras oncogene product, p21 and downstream target, the mitogen-activated protein kinases were significantly decreased by turmeric during HBP carcinogenesis. Turmeric also diminished the DMBA-induced mRNA expression of proto-oncogenes (c-jun, c-fos) and NF-kappaB, leading to decreased protein levels and in further attenuation of DMBA-induced AP-1/NF-kappaB DNA-binding in the buccal pouch nuclear extracts. Besides, buccal pouch of hamsters receiving turmeric diet showed significant alterations in DMBA-induced effects: (a) decrease in cell proliferation (diminished PCNA and Bcl2 expression), (b) enhanced apoptosis (increased expression of Bax, caspase-3 and apoptotic index), (c) decrease in inflammation (levels of Cox-2, the downstream target of AP-1/NF-kappaB, and PGE2) and (d) aberrant expression of differentiation markers, the cytokeratins (1, 5, 8, and 18). Together, the protective effects of dietary turmeric converge on augmenting apoptosis of the initiated cells and decreasing cell proliferation in DMBA-treated animals, which in turn, is reflected in decreased tumor burden, multiplicity and enhanced latency period. Some of these biomarkers are likely to be helpful in monitoring clinical trials and evaluating drug effect measurements.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/toxicidad , Curcuma , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias de la Boca/prevención & control , FN-kappa B/fisiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Factor de Transcripción AP-1/fisiología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Mejilla , Cricetinae , Dieta , Queratinas/análisis , Masculino , Mesocricetus , Proteínas Proto-Oncogénicas c-bcl-2/análisis
18.
Oncogene ; 37(34): 4735-4749, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29765153

RESUMEN

The pro-oncogenic kinase PKCε is overexpressed in human prostate cancer and cooperates with loss of the tumor suppressor Pten for the development of prostatic adenocarcinoma. However, the effectors driving PKCε-mediated phenotypes remain poorly defined. Here, using cellular and mouse models, we showed that PKCε overexpression acts synergistically with Pten loss to promote NF-κB activation and induce cyclooxygenase-2 (COX-2) expression, phenotypic traits which are also observed in human prostate tumors. Targeted disruption of PKCε from prostate cancer cells impaired COX-2 induction and PGE2 production. Notably, COX-2 inhibitors selectively killed prostate epithelial cells overexpressing PKCε, and this ability was greatly enhanced by Pten loss. Long-term COX-2 inhibition markedly reduced adenocarcinoma formation, as well as angiogenesis in a mouse model of prostate-specific PKCε expression and Pten loss. Overall, our results provide strong evidence for the involvement of the canonical NF-κB pathway and its target gene COX2 as PKCε effectors, and highlight the potential of PKCε as a useful biomarker for the use of COX inhibition for chemopreventive and/or chemotherapeutic purposes in prostate cancer.


Asunto(s)
Carcinogénesis/genética , Ciclooxigenasa 2/genética , Neoplasias de la Próstata/genética , Proteína Quinasa C-epsilon/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , FN-kappa B/genética , Próstata/patología , Neoplasias de la Próstata/patología , Transducción de Señal/genética
19.
Clin Imaging ; 31(5): 321-4, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17825739

RESUMEN

We present three cases of malignant superior vena cava obstruction demonstrating infraphrenic venous collaterals, one of which showed intense focal hepatic enhancement on computed tomographic scan. Infraphrenic venous collaterals and focal liver enhancement are uncommon but specific computed tomographic features of superior vena cava obstruction.


Asunto(s)
Síndrome de la Vena Cava Superior/diagnóstico por imagen , Síndrome de la Vena Cava Superior/etiología , Neoplasias Torácicas/complicaciones , Neoplasias Torácicas/diagnóstico por imagen , Vena Cava Superior/diagnóstico por imagen , Adolescente , Adulto , Anciano , Humanos , Masculino , Radiografía
20.
J Clin Biochem Nutr ; 40(2): 82-91, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18188409

RESUMEN

Cancer chemoprevention is fast becoming a lucrative approach for controlling cancer. Carcinogenesis being a complex multi-step, multi-factorial process, a number of chemopreventive interventions can be employed. These strategies are generally directed against two broad events of carcinogenesis viz., initiation and promotion/progression. Anti-initiation interventions principally involve inhibition of carcinogen activation, scavenging of free radicals and reactive carcinogen metabolites along with enhanced detoxification of carcinogens by modulating cellular metabolism. Anti-promotion strategies involve attenuation of enhanced cellular proliferation along with induction of cellular apoptosis and differentiation. Dietary agents or herbal anti-oxidants due to low toxicity and relative safety are promising chemopreventive agents. These agents after emerging successful through a series of in vitro and in vivo assays enter clinical trials. Many dietary compounds have emerged as promising chemopreventive agents in empirical experiments. However, in clinical trials these compounds have met with limited success. This emphasizes the need for further detailed research on the mechanisms of observed chemoprevention and choice, dose, duration and bioavailability of chemopreventive agent used. Complex issues such as choice and nutritional status of target population, genetic variation, gene-environment interactions and relevance of biomarkers analyzed also warrant further research and analyses.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA