Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Cell Biochem ; 476(2): 743-755, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33070276

RESUMO

Statins are potent inhibitors of the mevalonate/cholesterol biosynthetic pathway and are widely prescribed for the prevention of cardiovascular diseases. Here, we carried out a comprehensive analysis of the effects of three statins, simvastatin, atorvastatin, and lovastatin, on six different cancer cell lines that include a P-glycoprotein-expressing, multidrug resistant variant of an ovarian cancer cell line. Incubation of all cancer cell lines with statins resulted in suppression of cell proliferation without inducing apoptotic cell death. The cell proliferation arrest could be reversed upon transfer of cells to statin-free growth media as well as by the supplementation of the growth media with mevalonate. Further analysis suggested that statins induced cell cycle arrest at G0/G1 phase in four cancer cell lines and the loss of c-Myc protein in three cancer cell lines. The c-Myc expression and the progression of cell division cycle were restored upon the addition of mevalonate to the culture media containing statins. Finally, cells incubated with statins contained an increased level of phosphorylated histone H2AX, an observation previously correlated to cellular senescence. Together, these data demonstrate that statins inhibit the mevalonate pathway which is tightly coupled to oxidative branch of the pentose phosphate pathway, c-Myc expression, cell division cycle progression, and cellular senescence. Implications of these observations in the application of statins as cancer therapeutics are discussed.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Atorvastatina/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Fluvastatina/farmacologia , Humanos , Lovastatina/farmacologia , Ácido Mevalônico/metabolismo , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/biossíntese , Sinvastatina/farmacologia
2.
Pharmacogenomics J ; 18(4): 601-608, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29520079

RESUMO

Comparison of human genome sequences from different individuals has unraveled that genes involved in the drug efficacy and metabolism are polymorphic, harboring mutations, splicing variations and other alterations. These data provide a reasonable explanation for the inter-individual variations observed in drug therapy. Thus, a detailed molecular analysis and an in-depth knowledge of these genes is a prerequisite to practice pharmacogenomics-based medicine. We have introduced a 6-week laboratory research rotation to train students in the expression analysis of different pharmacogenes combined with bioinformatics tools. Students were first introduced to the bioinformatics tools to identify appropriate DNA primers to amplify specific pharmacogenes from the laboratory cancer cell lines. The amplified DNA fragments were sequenced. Finally, students were trained in bioinformatics tools to establish the identity of these DNA sequences. The possible implications of this laboratory training in developing problem-solving skills needed in the implementation of pharmacogenomics knowledge in the clinic, are discussed.


Assuntos
Educação em Farmácia , Farmacogenética/educação , Medicina de Precisão/tendências , Estudantes de Farmácia , Humanos , Pesquisa/educação
3.
Tumour Biol ; 39(3): 1010428317691687, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28345468

RESUMO

One long-term complication of chronic intestinal inflammation is the development of colorectal cancer. However, the mechanisms linking inflammation to the colorectal tumorigenesis are poorly defined. Previously, we have demonstrated that galectin-4 is predominantly expressed in the luminal epithelia of the gastrointestinal tract, and its loss of expression plays a key role in the colorectal tumorigenesis. However, the mechanism by which galectin-4 regulates inflammation-induced tumorigenesis is unclear. Here, we show that galectin-4 secreted by the colorectal cancer cell lines was bound to the cell surface. Neutralization of surface-bound galectin-4 with anti-galectin-4 antibody resulted in increased cell proliferation with concomitant secretion of several chemokines into the extracellular medium. Neutralization of the surface-bound galectin-4 also resulted in the up-regulation of transcription of 29 genes, several of which are components of multiple inflammation signaling pathways. In an alternate experiment, binding of recombinant galectin-4 protein to cell surface of the galectin-4-negative colorectal cancer cells resulted in increased p27, and decreased cyclin D1 and c-Myc levels, leading to cell cycle arrest and apoptosis. Together, these data demonstrated that surface-bound galectin-4 is a dual function protein-down-regulating cell proliferation and chemokine secretion in galectin-4-expressing colorectal cancer cells on one hand and inducing apoptosis in galectin-4-negative colorectal cancer cells on the other hand.


Assuntos
Carcinogênese/patologia , Quimiocinas/metabolismo , Neoplasias Colorretais/patologia , Galectina 4/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Mucosa Intestinal/patologia , Apoptose/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Quimiocinas/biossíntese , Neoplasias Colorretais/genética , Ciclina D1/metabolismo , Células HCT116 , Células HT29 , Humanos , Inflamação/patologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo
4.
Tumour Biol ; 35(7): 7267-73, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24777335

RESUMO

Assessment of the human epidermal growth factor receptor-2 (Her2/ErbB2) and estrogen receptor (ER) and progesterone receptor (PR) expression in breast cancer has been an accepted standard to predict clinical outcome. Expression of these receptors in primary breast cancer has also been an important predictor of visceral organ metastasis. Many studies of breast cancer have reported risk factors for brain metastasis that include Her2/ErbB2 positivity, ER negativity, and negativity for all the above three receptors. However, it is not clear whether expression of these receptors would persist subsequent to brain metastasis. To address this possibility, we analyzed different breast cancer brain metastases (BCBM) for the expression of Her2/ErbB2, ER, and PR by immunohistochemistry procedure. The results showed that BCBM are heterogeneous in the receptor expression: Five BCBMs were Her2/ErbB2-positive and one negative; four BCBMs were ER-positive, and two were negative; five BCBMs were PR-positive and one negative. However, expression of these receptors in their combination is also heterogeneous: Four BCBMs were positive for all of the Her2/ErbB2, ER, and PR; one BCBM was positive for Her2/ErbB2 and PR but negative for ER; one BCBM was positive for PR but negative for Her2/ErbB2/ER. Similar heterogeneity in the expression of these receptors was also observed in primary tumors. Importantly, BCBM tumors that were assigned as ER- and PR-positive contained tumor cells that lacked expression of these receptors in other regions of the biopsies. Taken together, our findings indicate that the BCBM exhibit heterogeneity in the expression amounts of Her2/ErbB2, ER, and PR, which could be a result of the influence of tumor microenvironment in the brain or different tumor cells populating the metastatic region.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias da Mama/genética , Receptor alfa de Estrogênio/biossíntese , Receptor ErbB-2/biossíntese , Receptores de Progesterona/biossíntese , Biomarcadores Tumorais/biossíntese , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Receptor ErbB-2/genética , Receptores de Progesterona/genética
5.
J Gastroenterol Hepatol ; 28(3): 505-12, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23216017

RESUMO

BACKGROUND AND AIM: The usefulness of carcinoembryonic antigen (CEA) in the diagnosis and prognosis of colorectal cancer (CRC) is unclear. The aim was to analyze changes in the expression of CEA during CRC progression and metastasis, so as to determine the influence of tumor metastatic organ on the CEA expression by CRC cells. METHODS: The human biopsies of adenocarcinomas in colon and CRC liver and lung metastases were analyzed by immunohistochemistry for the expression of CEA. Expression of E-cadherin and ß-catenin was also analyzed to localize the CRC neoplastic glands in metastatic tissues. RESULTS: The CRC neoplastic glands in colon and liver expressed significantly higher amount of CEA compared with crypts in normal colon. In contrast, CRC neoplastic glands formed in lung expressed low CEA level. However, CEA expression was high in areas of tumor necrosis in lung. E-cadherin and ß-catenin were cell membrane-bound in normal crypts and CRC neoplastic glands in colon and liver. Although these two proteins were also cell membrane-bound in a majority of CRC neoplastic glands in lungs, a significant proportion of these expressed ß-catenin in the nucleus, which lacked either E-cadherin or ß-catenin at the cell membrane. CONCLUSION: Our findings indicate that lung microenvironment is unique in that it suppresses the expression of CEA by CRC cells forming neoplastic glands. In addition, lung microenvironment promotes nuclear localization of ß-catenin, suggesting that the Wnt signaling pathway is relatively active highly in CRC metastasized to lung, when compared with liver or colon.


Assuntos
Adenocarcinoma/secundário , Caderinas/metabolismo , Antígeno Carcinoembrionário/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , beta Catenina/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Biópsia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/metabolismo , Humanos , Imuno-Histoquímica , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia
6.
Int J Cancer ; 130(11): 2703-14, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21792893

RESUMO

Bioflavonoids are of considerable interest to human health as these serve as antioxidant and anticancer agents. Although epidemiological and experimental studies suggest that luteolin, a natural bioflavonoid, exhibits chemopreventive properties, its effectiveness as an antiproliferative agent against multidrug resistant (MDR) cancers is unclear. Thus, we assessed the antiproliferative effects of luteolin and associated molecular mechanisms using two MDR cancer cell lines that express high levels of P-glycoprotein and ABCG2. In this article, we demonstrate that luteolin induces apoptosis in P-glycoprotein- and ABCG2-expressing MDR cancer cells without affecting the transport functions of these drug transporters. Analysis of various proliferative signaling pathways indicated that luteolin-induced apoptosis involves reactive oxygen species generation, DNA damage, activation of ATR → Chk2 → p53 signaling pathway, inhibition of NF-kB signaling pathway, activation of p38 pathway and depletion of antiapoptotic proteins. Importantly, use of luteolin in these analyses also identified specific molecular characteristics of NCI-ADR/RES and MCF-7/Mito(R) cells that highlight their different tissue origins. These results suggest that luteolin possesses therapeutic potential to control the proliferation of MDR cancers without affecting the physiological function of drug transporters in the body tissues.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Luteolina/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/fisiologia , Caspase 7/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Neoplasias/fisiologia , Espécies Reativas de Oxigênio/metabolismo
7.
Int J Cancer ; 129(4): 799-809, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21064109

RESUMO

Development of colorectal cancer (CRC) involves a series of genetic alterations with altered expression of proteins and cell signaling pathways. Here, we identified that galectin-4 (gal-4), a marker of differentiation, was down-regulated in CRC. The goal of this work was to determine the function of gal-4 in CRC. Toward this goal, the human colon biopsies and tissue microarrays containing a gradient of pathology were analyzed for gal-4 expression by immunohistochemistry. Cell proliferation, migration, motility, forced expression, knockdown, cell cycle and apoptosis assays were used to characterize gal-4 function. Immunohistochemistry identified that gal-4 expression was significantly down-regulated in adenomas and was essentially absent in invasive carcinomas. Forced expression of gal-4 in gal-4 -ve cells induced cell cycle arrest and retarded cell migration and motility. Further, gal-4 sensitized the cells to camptothecin-induced apoptosis. Gal-4 knockdown resulted in increased cell proliferation, migration and motility. Gal-4 was found to be associated with Wnt signaling proteins. Finally, gal-4 expression led to down-regulation of Wnt signaling target genes. This study demonstrates that loss of gal-4 is a common and specific event in CRC. This study also shows that gal-4 exhibits tumor suppressive effects in CRC cells in vitro. Through its ability to interact with and down-regulate the functions of Wnt signaling pathway, gal-4 reveals a new dimension in the control of the Wnt signaling pathway. Thus, gal-4 may prove to be an important molecule in understanding the biology of CRC.


Assuntos
Adenocarcinoma/metabolismo , Adenoma/metabolismo , Neoplasias Colorretais/metabolismo , Galectina 4/metabolismo , Genes Supressores de Tumor , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenoma/genética , Adenoma/patologia , Apoptose , Western Blotting , Adesão Celular , Ciclo Celular , Movimento Celular , Proliferação de Células , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação para Baixo , Galectina 4/antagonistas & inibidores , Galectina 4/genética , Humanos , Técnicas Imunoenzimáticas , Invasividade Neoplásica , RNA Interferente Pequeno/genética , Transdução de Sinais , Células Tumorais Cultivadas , Proteínas Wnt/metabolismo , Cicatrização , beta Catenina/metabolismo
8.
Proteomics ; 9(10): 2776-87, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19405034

RESUMO

RNF2, a member of polycomb group (PcG) proteins, is involved in chromatin remodeling. However, mechanisms that regulate RNF2 function are unknown. To identify such mechanisms, RNF2 was expressed in HEK-293 cells and analyzed by 2-D electrophoresis. RNF2 was resolved into at least seven protein spots, migrating toward the lower pI from its expected pI of 6.38, suggesting that RNF2 undergoes post-translational modifications. Western blotting indicated that majority of these RNF2 spots contained phosphoserine(s), which were completely dephosphorylated upon treatment with a phosphatase. SB203580, a specific inhibitor of p38 MAPK, inhibited RNF2 phosphorylation at one site. On the other hand, PD98059, an inhibitor of MEK1/2, inhibited majority of the phosphorylation events in RNF2. Mass spectrometry analysis identified that RNF2 expressed in Sf9 insect cells undergoes co-translational excision of (1)Met coupled to N-acetylation of (2)Ser, and phosphorylation of (41)Ser. Interestingly, (41)Ser is a predicted p38/MAPK phosphorylation site, consistent with the loss of phosphorylation induced by SB203580. Further analysis indicated that RNF2 phosphorylation differentially modulates the expression of transcription factors and histone 2B acetylation. These results provide first evidence for phosphorylation of RNF2, and suggest that the mitogen activated protein kinases including p38 MAPK and ERK1/2 regulate growth, stress response, differentiation and other cellular processes, through phosphorylation of RNF2.


Assuntos
Proteínas de Ligação a DNA/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Repressoras/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/genética , Eletroforese em Gel Bidimensional , Regulação da Expressão Gênica , Humanos , Fosforilação , Fosfosserina/metabolismo , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Processamento de Proteína Pós-Traducional , Proteômica , Proteínas Repressoras/genética , Espectrometria de Massas em Tandem , Ubiquitina-Proteína Ligases/genética
9.
J Gen Physiol ; 130(6): 611-29, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17998393

RESUMO

The epithelial Na(+) channel (ENaC) that mediates regulated Na(+) reabsorption by epithelial cells in the kidney and lungs can be activated by endogenous proteases such as channel activating protease 1 and exogenous proteases such as trypsin and neutrophil elastase (NE). The mechanism by which exogenous proteases activate the channel is unknown. To test the hypothesis that residues on ENaC mediate protease-dependent channel activation wild-type and mutant ENaC were stably expressed in the FRT epithelial cell line using a tripromoter human ENaC construct, and protease-induced short-circuit current activation was measured in aprotinin-treated cells. The amiloride-sensitive short circuit current (I(Na)) was stimulated by aldosterone (1.5-fold) and dexamethasone (8-fold). Dexamethasone-treated cells were used for all subsequent studies. The serum protease inhibitor aprotinin decreased baseline I(Na) by approximately 50% and I(Na) could be restored to baseline control values by the exogenous addition of trypsin, NE, and porcine pancreatic elastase (PE) but not by thrombin. All protease experiments were thus performed after exposure to aprotinin. Because NE recognition of substrates occurs with a preference for binding valines at the active site, several valines in the extracellular loops of alpha and gamma ENaC were sequentially substituted with glycines. This scan yielded two valine residues in gamma ENaC at positions 182 and 193 that resulted in inhibited responses to NE when simultaneously changed to other amino acids. The mutations resulted in decreased rates of activation and decreased activated steady-state current levels. There was an approximately 20-fold difference in activation efficiency of NE against wild-type ENaC compared to a mutant with glycine substitutions at positions 182 and 193. However, the mutants remain susceptible to activation by trypsin and the related elastase, PE. Alanine is the preferred P(1) position residue for PE and substitution of alanine 190 in the gamma subunit eliminated I(Na) activation by PE. Further, substitution with a novel thrombin consensus sequence (LVPRG) beginning at residue 186 in the gamma subunit (gamma(Th)) allowed for I(Na) activation by thrombin, whereas wild-type ENaC was unresponsive. MALDI-TOF mass spectrometric evaluation of proteolytic digests of a 23-mer peptide encompassing the identified residues (T(176)-S(198)) showed that hydrolysis occurred between residues V193 and M194 for NE and between A190 and S191 for PE. In vitro translation studies demonstrated thrombin cleaved the gamma(Th) but not the wild-type gamma subunit. These results demonstrate that gamma subunit valines 182 and 193 are critical for channel activation by NE, alanine 190 is critical for channel activation by PE, and that channel activation can be achieved by inserting a novel thrombin consensus sequence. These results support the conclusion that protease binding and perhaps cleavage of the gamma subunit results in ENaC activation.


Assuntos
Canais Epiteliais de Sódio/fisiologia , Elastase de Leucócito/metabolismo , Sódio/metabolismo , Corticosteroides/farmacologia , Algoritmos , Sequência de Aminoácidos , Animais , Transporte Biológico Ativo , Western Blotting , Ativação Enzimática/genética , Ativação Enzimática/fisiologia , Canais Epiteliais de Sódio/genética , Humanos , Elastase de Leucócito/genética , Dados de Sequência Molecular , Elastase Pancreática/genética , Elastase Pancreática/metabolismo , Peptídeo Hidrolases/fisiologia , Regiões Promotoras Genéticas/genética , RNA/biossíntese , RNA/genética , RNA/isolamento & purificação , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Suínos , Trombina/metabolismo , Transfecção
10.
Int J Oncol ; 32(1): 135-44, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18097552

RESUMO

P-glycoprotein (Pgp), a plasma membrane (PM) glycoprotein, is responsible for the development of multidrug resistance. The mechanism by which Pgp is targeted to the PM is not defined. To identify proteins that influence Pgp trafficking, we utilized the yeast two-hybrid analysis procedure, which identified a new isoform of endoplasmic reticulum (ER)-bound Bap29, termed Bap29varP, as an interacting protein with the N-terminus of Pgp. The drug-resistant human breast cancer MCF-7 (MCF-7/Adr(R)) cells express both Bap29varP and approximately 170 kDa Pgp, which are however absent in the drug-sensitive MCF-7 cells. When Bap29varP was overexpressed in MCF-7/Adr(R) cells, Pgp was predominantly localized in the ER and intracellular vesicles, suggesting Bap29varP influences Pgp trafficking. When Pgp was expressed in MCF-7 cells, it was exclusively found in the ER with a molecular mass of approximately 160 kDa slightly smaller than that of the molecular mass of Pgp expressed in MCF-7/Adr(R) cells. On the other hand, when Pgp was expressed in Bap29varP-containing human colon adenocarcinoma HT-29 cells, it was localized at the PM. These findings together suggest that Bap29varP acts as an essential chaperone, influencing the processing and trafficking of Pgp to the cell surface.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/análise , Proteínas de Membrana/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/metabolismo , Glicosilação , Humanos , Proteínas de Membrana/análise , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Isoformas de Proteínas , Processamento de Proteína Pós-Traducional , Receptores de Estrogênio/análise
11.
Oncol Rep ; 19(3): 587-94, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18288388

RESUMO

Galectins play a key role in oncogenic processes. Although several galectins are known, their relative expression at the mRNA and protein levels, the subcellular localization, and their relationship to the oncogenic manifestation remains unclear. Herein we report a comprehensive characterization of the expression of major galectins in human breast cancer (drug-sensitive MCF-7 and drug-resistant MCF-7/Adr(R)), colon cancer (HCT-116 and HT-29), and glioma (T98G) cell lines, as these cells are common model systems for studying oncogenic processes. The expected approximately 14.5 kDa galectin-1, predominantly cytosolic, was detected in the cancer and normal cell lines. Notably, two different molecular forms of galectin-1 with molecular masses of approximately 13.5 and 15 kDa were detected in T98G cells, the latter being in the extracellular medium, perhaps a result of post-translational processing. Immunocytochemistry indicated that the extracellular galectin-1 bound to the cell surface was punctated in appearance, suggesting that it was bound to specific receptors. Immunohistological studies indicated that metastasizing carcinomas express high levels of galectin-1. On the other hand, galectin-3 was readily detectable in all cancer cell lines but undetectable in normal cell lines, indicating that galectin-3 is a cancer-specific biomarker protein. Galectin-3 was a cytosolic protein but was not detected in the extracellular medium, indicating that cancer cells do not secrete this galectin. Finally, despite the RT-PCR analysis suggesting the presence of two transcripts of galectin-8 in all cancer cell lines, the corresponding approximately 36 kDa protein was only detectable in the nuclear and cytosolic fractions upon cell fractionation. Notably, a different molecular form of galectin-8 of approximately 18 kDa was immunoprecipitated from the extracellular media, suggesting that the secreted galectin-8 undergoes post-translational processing. These results highlight the expression of galectins in different molecular forms in cancers, warranting caution in interpreting the results of functional studies of individual galectins, particularly because these proteins function redundantly in cancer pathways.


Assuntos
Galectinas/análise , Galectinas/metabolismo , Neoplasias/metabolismo , Neoplasias da Mama/química , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Neoplasias do Colo/química , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Galectina 1/química , Galectina 1/genética , Galectina 1/metabolismo , Galectina 3/química , Galectina 3/genética , Galectina 3/metabolismo , Galectinas/química , Galectinas/genética , Humanos , Imuno-Histoquímica , Neoplasias/química , Neoplasias/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Int J Oncol ; 29(5): 1269-78, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17016661

RESUMO

O6-alkylguanines are potent mutagenic, pro-carcinogenic and cytotoxic lesions induced by exogenous and endogenous alkylating agents. A facilitated elimination of these lesions by increasing the activity of O6-methylguanine-DNA methyltransferase (MGMT) is likely to be a beneficial chemoprevention strategy, which, however, has not been examined. Because, a marginal enhancement of this protein may be adequate for genomic protection, we studied alterations in MGMT activity and expression in human peripheral blood lymphocytes and cancer cell lines induced by water-soluble and alcohol-soluble constituents of several plants with established antioxidant and medicinal properties. Both the ethanolic and aqueous extracts from neem (Azadirachta indica), holy basil (Ocimum sanctum), winter cherry (Withania somnifera), and oregano (Origanum majorana) increased the levels of MGMT protein and its demethylation activity in a time-dependent manner with a maximum of 3-fold increase after 72-h treatment. The extracts from gooseberry (Emblica officinalis), common basil (Ocimum basilicum), and spearmint (Mentha viridis) were relatively less efficient in raising MGMT levels. Increased levels of MGMT mRNA accounted at least, in part, for the increased activity of the DNA repair protein. The herbal treatments also increased glutathione S-transferase-pi (GSTP1) expression, albeit to a lesser extent than MGMT. These data provide the first evidence for the upregulation of human MGMT by plant constituents and raise the possibility of rational dietary approaches for attenuating alkylation-induced carcinogenesis. Further, they reveal the putative antioxidant responsiveness of the MGMT gene in human cells.


Assuntos
Metilação de DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Magnoliopsida/química , Neoplasias/prevenção & controle , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Extratos Vegetais/uso terapêutico , Linhagem Celular Tumoral , Quimioprevenção , Etanol/química , Expressão Gênica/efeitos dos fármacos , Glutationa S-Transferase pi/genética , Glutationa S-Transferase pi/metabolismo , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/enzimologia , Neoplasias/enzimologia , Neoplasias/genética , O(6)-Metilguanina-DNA Metiltransferase/análise , O(6)-Metilguanina-DNA Metiltransferase/genética , Extratos Vegetais/química , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Água/química
13.
Int J Oncol ; 29(6): 1413-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17088979

RESUMO

The human P-glycoprotein (Pgp) is a drug-efflux pump responsible for innate or acquired multidrug resistance in many cancers. Pgp contains a unique approximately 75 amino acid long linker region in its middle, which is critically important for its drug transport and ATPase functions. To identify cellular proteins that bind to this linker region and modulate Pgp function, a yeast two-hybrid analysis was carried out. This procedure identified RNF2 (RING finger protein 2), an E3 ubiquitin ligase, as a prominent Pgp-interacting protein. Co-expression of RNF2 with Pgp in Sf9 insect cells resulted in decreased ATPase activity and proteolytic protection of the transporter protein. Immunoprecipitation experiments confirmed the physical interaction between these two proteins. Confocal microscopy showed the presence of RNF2 in the cytoplasm of the Pgp-negative, drug-sensitive MCF-7 breast cancer cells. However, it was undetectable in the Pgp-positive and drug-resistant MCF-7 cells. We suggest that RNF2 regulates the cellular abundance of Pgp, and plays a key role in the development of cancer drug resistance through its own down-regulation.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adenocarcinoma/metabolismo , Neoplasias da Mama/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Adenocarcinoma/genética , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Linhagem Celular Tumoral , DNA Complementar/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Complexo Repressor Polycomb 1 , Ligação Proteica , Spodoptera/virologia , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/genética , Leveduras/genética
14.
Clin Cancer Res ; 11(16): 5833-9, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16115923

RESUMO

PURPOSE: Cancers exposed to chemotherapy develop multidrug resistance, a major cause for chemotherapy failure. One mechanism of multidrug resistance development is due to overexpression of P-glycoprotein (Pgp) in these cancer cells. Thus, a prechemotherapy evaluation of Pgp in cancer cells aids in the design of alternative regimens that can circumvent such failure. As few Pgp-specific antibodies are available in detecting low levels of Pgp, there is a need for preparing an antibody that allows the detection of Pgp by various immunologic methods. EXPERIMENTAL DESIGN: We selected the amino acid stretch 11 to 34 in the cytoplasmically located NH2 terminus of Pgp as antigen, which was chemically synthesized and used to raise an antibody in a rabbit, termed NH2 11 antibody. We compared the properties of NH2 11 antibody with that of the well-characterized Pgp-specific antibody, C219, by Western blotting, immunoprecipitation, immunocytochemistry, and immunohistochemistry. RESULTS: Immunoblotting analysis suggested that NH2 11 antibody efficiently interacts with both recombinant and constitutively expressed Pgp in cancerous and noncancerous human cells. Immunoprecipitation reactions indicated that the NH2 11 antibody selectively immunoprecipitates Pgp. Immunocytochemical analyses indicated that the NH2 11 antibody detects Pgp in drug-resistant breast cancer cells as well as in human prostate and breast adenocarcinoma tissue sections. CONCLUSION: As the NH2 11 antibody detects Pgp present in cells and tissues, we conclude that the amino acid sequence to which this antibody was raised is highly antigenic and the antibody is useful in the detection of Pgp by a variety of immunologic methods.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/imunologia , Anticorpos/imunologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/química , Sequência de Aminoácidos , Anticorpos/análise , Especificidade de Anticorpos , Western Blotting , Neoplasias da Mama/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Epitopos/imunologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Dados de Sequência Molecular , Fragmentos de Peptídeos/imunologia , Neoplasias da Próstata/imunologia
15.
Pharmacogenomics ; 16(8): 905-11, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26067487

RESUMO

Sequencing of the human genome and the evidence correlating specific genetic variations to diseases have opened up the potential of genomics to more effective and less harmful interventions of human diseases. A wealth of pharmacogenomics knowledge is in place for the practice of precision medicine. However, this knowledge is not fully realized in clinical practice. One reason for this impasse is the lack of in-depth understanding of the potential of pharmacogenomics among the healthcare professionals. Pharmacists are the point-of-care providers and are expected to advise clinicians on matters relating to the implementation of pharmacogenomics in patient care. However, current pharmacogenomics instruction in pharmacy schools fails to produce pharmacists with the required knowledge or practical training in this discipline. In this perspective, we provide several strategies to overcome limitations faced by pharmacy schools. Once implemented, pharmacy schools will produce precision medicine-ready pharmacists.


Assuntos
Educação em Farmácia , Farmacogenética/educação , Currículo , Genoma Humano , Genômica/educação , Humanos , Faculdades de Farmácia
16.
Cell Calcium ; 35(1): 21-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14670368

RESUMO

The amiloride-sensitive epithelial sodium channel (ENaC), a multimeric plasma membrane protein composed of alpha-, beta-, and gamma-ENaC subunits, mediates Na(+) reabsorption in epithelial tissues, including the distal nephron, colon, lung, and secretory glands, and plays a critical role in pathophysiology of essential hypertension and cystic fibrosis (CF). The function of ENaC is tightly regulated by signals elicited by aldosterone, vasopressin, agents that increase intracellular cAMP levels, ions, ion channels, G-protein-coupled mechanisms, and cytoskeletal proteins. In this paper, the effects of Ca(2+) on the expression of the human ENaC subunits expressed in human embryonic kidney cells (HEK-293 cells) were examined. Incubation of cells with increased extracellular Ca(2+) and treatment of cells with A23187 and thapsigargin stimulated the expression of the monomeric ENaC subunits. Treatment of cells with Ca(2+)-chelating agents, EGTA and BAPTA-AM, reduced the levels of ENaC subunit expression. The pulse-chase experiments suggested that a rise in the intracellular Ca(2+) increases the ENaC subunit expression. Immunoblot analysis using the anti-ubiquitin antibody indicated that ENaC undergoes ubiquitination. A correlation between the processes that regulate ENaC function with the intracellular Ca(2+) was discussed.


Assuntos
Cálcio/fisiologia , Canais de Sódio/biossíntese , Sequência de Aminoácidos , Calcimicina/farmacologia , Linhagem Celular , Clonagem Molecular , Citosol/metabolismo , Canais Epiteliais de Sódio , Humanos , Rim/citologia , Dados de Sequência Molecular , Peso Molecular , Subunidades Proteicas/biossíntese , Proteínas Recombinantes/biossíntese , Tapsigargina/farmacologia
17.
Cancer Lett ; 301(1): 38-46, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21122983

RESUMO

Galectin-1 (gal-1) is an important molecule secreted by many tumors, which induces apoptosis in activated T-cells and promotes tumor angiogenesis, both of which phenomena facilitate successful establishment of tumor in the body. However, little is known about the function of intracellular gal-1 or its transcriptional regulation in colorectal cancer (CRC). Here, we demonstrate that gal-1 expression is epigenetically regulated in CRC through promoter hypermethylation. Intracellular gal-1 induces cell cycle arrest and apoptosis in CRC cells with concomitant down-regulation of Wnt and NF-κB signaling pathways. Together, these data suggested that gal-1 silencing imparts CRC with the ability to proliferate and escape apoptosis.


Assuntos
Apoptose , Neoplasias Colorretais/etiologia , Metilação de DNA , Galectina 1/fisiologia , Inativação Gênica , Regiões Promotoras Genéticas , Apoptose/efeitos dos fármacos , Butiratos/farmacologia , Caspases/fisiologia , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Galectina 1/antagonistas & inibidores , Galectina 1/genética , Humanos , Potencial da Membrana Mitocondrial
19.
J Biol Chem ; 277(33): 29417-23, 2002 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-12055198

RESUMO

P-glycoprotein (Pgp), an anticancer drug-translocating ATPase, is responsible for multidrug resistance in cancer. We have previously shown (Nuti, S. L., Mehdi, A., and Rao, U. S. (2000) Biochemistry 39, 3424-3432) that tryptic cleavage of Pgp results in the activation of basal and drug-stimulated ATPase functions of Pgp. To understand this phenomenon, we determined the sites cleaved by trypsin and further examined whether the modulation of Pgp function is trypsin-specific or the result of proteolysis in general. The effects of chymotrypsin and proteinase K on Pgp ATPase function were studied. The results show that proteolysis of Pgp irrespective of the protease employed resulted in the activation of basal ATPase activity. However, drug-stimulated ATPase activities were differentially modulated. Immunoblot analysis of proteolytic digests indicated that, irrespective of the protease employed, Pgp was predominantly cleaved in the middle of the molecule. N-terminal amino acid sequencing of Pgp tryptic and chymotryptic peptides indicated Arg(680) and Leu(682) as the sites of cleavage, respectively. These two cleavage sites are part of the predicted linker region that joins the two halves of Pgp. Together, these results suggest that the linker region in Pgp is primarily accessible to protease action and that cleavage of this region modulates Pgp ATPase function.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Humanos , Hidrólise , Mutação , Spodoptera
20.
J Biol Chem ; 278(47): 46576-82, 2003 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-12968025

RESUMO

P-glycoprotein (Pgp) is a drug-translocating ATPase responsible for multidrug resistance in cancer. Although it is well-established that Pgp exhibits drug-dependent ATPase and ATP-dependent drug transport functions, the mechanism by which these two reactions are coupled remains unclear. We have shown recently that proteolytic cleavage of the linker region, which joins the NH2 and COOH halves of the Pgp molecule, results in a Pgp form that exhibits drug-independent and -dependent ATPase activities (Nuti et al., (2000) Biochemistry 39, 3424-3432; Nuti, S. L., and Rao, U. S. (2002) J. Biol. Chem. 277, 29417-29423). To understand the mechanism underlying this phenomenon, we used the procedure of vanadate-mediated trapping of the Pgp transport cycle intermediates to determine the steps in the catalytic cycle that are being regulated by the linker region. We show that vanadate stably traps Pgp under two different conditions, one in the presence of ATP alone and the other in the presence of ATP and drug, suggesting the existence of two Pgp conformations. These two conformations, one mediating basal and the other drug-stimulated ATPase reactions, represent different transport cycle intermediates of Pgp, because arresting Pgp in either conformation prevents the catalytic cycle from proceeding to completion. The results also show that these two conformations are uncoupled and appear simultaneously in Pgp that was cleaved in the linker region. These results together suggest that Pgp assumes at least two distinct conformational states, which catalyze two ATP hydrolysis events in the drug transport cycle, and the linker region mediates the transition between these two states of Pgp.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/química , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adenosina Trifosfatases , Trifosfato de Adenosina/farmacologia , Sítios de Ligação , Transporte Biológico , Catálise , Humanos , Modelos Químicos , Conformação Proteica , Vanadatos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa