Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Adv Exp Med Biol ; 1408: 291-308, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37093434

RESUMO

Prostate-specific membrane antigen (PSMA) is expressed in epithelial cells of the prostate gland and is strongly upregulated in prostatic adenocarcinoma, with elevated expression correlating with metastasis, progression, and androgen independence. Because of its specificity, PSMA is a major target of prostate cancer therapy; however, detectable levels of PSMA are also found in other tissues, especially in salivary glands and kidney, generating bystander damage of these tissues. Antibody target therapy has been used with relative success in reducing tumor growth and prostate specific antigen (PSA) levels. However, since antibodies are highly stable in plasma, they have prolonged time in circulation and accumulate in organs with an affinity for antibodies such as bone marrow. For that reason, a second generation of PSMA targeted therapeutic agents has been developed. Small molecules and minibodies have had promising clinical trial results, but concerns about their specificity had arisen with side effects due to accumulation in salivary glands and kidneys. Herein we study the specificity of small molecules and minibodies that are currently being clinically tested. We observed a high affinity of these molecules for PSMA in prostate, kidney and salivary gland, suggesting that their effect is not prostate specific. The search for specific prostate target agents must continue so as to optimally treat patients with prostate cancer, while minimizing deleterious effects in other PSMA expressing tissues.


Assuntos
Neoplasias da Próstata , Masculino , Humanos , Neoplasias da Próstata/patologia , Antígenos de Superfície/metabolismo , Antígeno Prostático Específico
2.
Mol Carcinog ; 59(8): 955-966, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32391971

RESUMO

Maspin repression is frequently observed in prostate cancer; however, the molecular mechanism(s) causing the loss is not completely understood. Here, we demonstrate that inhibition of class I histone deacetylases (HDACs) mediates re-expression of maspin which plays an essential role in suppressing proliferation and migration capability in prostate cancer cells. Human prostate cancer LNCaP and DU145 cells treated with HDAC inhibitors, sodium butyrate, and trichostatin A, resulted in maspin re-expression. Interestingly, an exploration into the molecular mechanisms demonstrates that maspin repression in prostate tumor and human prostate cancer cell lines occurs via epigenetic silencing through an increase in HDAC activity/expression, independent of promoter DNA hypermethylation. Furthermore, transcriptional activation of maspin was accompanied with the suppression of HDAC1 and HDAC8 with significant p53 enrichment at the maspin promoter associated with an increase in histone H3/H4 acetylation. Our results provide evidence of maspin induction as a critical epigenetic event altered by class I HDACs in the restoration of balance to delay proliferation and migration ability of prostate cancer cells.


Assuntos
Biomarcadores Tumorais/metabolismo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/metabolismo , Neoplasias da Próstata/patologia , Serpinas/genética , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Epigênese Genética , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/química , Histona Desacetilases/genética , Histonas , Humanos , Ácidos Hidroxâmicos/farmacologia , Masculino , Prognóstico , Regiões Promotoras Genéticas , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Serpinas/metabolismo , Células Tumorais Cultivadas
3.
Mol Carcinog ; 53(1): 8-18, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22833520

RESUMO

The pi-class glutathione S-transferase (GSTP1) actively protect cells from carcinogens and electrophilic compounds. Loss of GSTP1 expression via promoter hypermethylation is the most common epigenetic alteration observed in human prostate cancer. Silencing of GSTP1 can increase generation of reactive oxygen species (ROS) and DNA damage in cells. In this study we investigated whether loss of GSTP1 contributes to increased DNA damage that may predispose men to a higher risk of prostate cancer. We found significantly elevated (103%; P < 0.0001) levels of 8-oxo-2'-deoxogunosine (8-OHdG), an oxidative DNA damage marker, in adenocarcinomas, compared to benign counterparts, which positively correlated (r = 0.2) with loss of GSTP1 activity (34%; P < 0.0001). Silencing of GSTP1 using siRNA approach in normal human prostate epithelial RWPE1 cells caused increased intracellular production of ROS and higher susceptibility of cells to H2 O2 -mediated oxidative stress. Additionally, human prostate carcinoma LNCaP cells, which contain a silenced GSTP1 gene, were genetically modified to constitutively express high levels of GSTP1. Induction of GSTP1 activity lowered endogenous ROS levels in LNCaP-pLPCX-GSTP1 cells, and when exposed to H2 O2 , these cells exhibited significantly reduced production of ROS and 8-OHdG levels, compared to vector control LNCaP-pLPCX cells. Furthermore, exposure of LNCaP cells to green tea polyphenols caused reexpression of GSTP1, which protected the cells from H2 O2 -mediated DNA damage through decreased ROS production compared to nonexposed cells. These results suggest that loss of GSTP1 expression in human prostate cells, a process that increases their susceptibility to oxidative stress-induced DNA damage, may be an important target for primary prevention of prostate cancer.


Assuntos
Dano ao DNA , Glutationa S-Transferase pi/metabolismo , Estresse Oxidativo , Próstata/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Catequina/análogos & derivados , Catequina/farmacologia , Linhagem Celular , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Ativação Enzimática , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutationa S-Transferase pi/genética , Guanosina Trifosfato/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Espécies Reativas de Oxigênio/metabolismo
4.
Int J Cancer ; 133(8): 1955-66, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23564295

RESUMO

Hedgehog (Hh) signaling is a highly conserved intercellular and intracellular communication mechanism that governs organogenesis and is dysregulated in cancers of numerous tissues, including prostate. Up-regulated expression of the Hh ligands, Sonic (Shh) and Desert (Dhh), has been reported in androgen-deprived and castration-resistant prostate cancer (CRPC). In a cohort of therapy naive, short- and long-term neoadjuvant hormone therapy-treated (NHT), and CRPC specimens, we observed elevated Dhh expression predominantly in long-term NHT specimens and elevated Shh expression predominantly in CRPC specimens. Together with previously demonstrated reciprocal signaling between Shh-producing prostate cancer (PCa) cells and urogenital mesenchymal fibroblasts, these results suggest that castration-induced Hh expression promotes CRPC progression through reciprocal paracrine signaling within the tumor microenvironment. We tested whether the orally available Smoothened (Smo) antagonist, TAK-441, could impair castration-resistant progression of LNCaP PCa xenografts by disrupting paracrine Hh signaling. Although TAK-441 or cyclopamine did not affect androgen withdrawal-induced Shh up-regulation or viability of LNCaP cells, castration-resistant progression of LNCaP xenografts was significantly delayed in animals treated with TAK-441. In TAK-441-treated xenografts, expression of murine orthologs of the Hh-activated genes, Gli1, Gli2 and Ptch1, was substantially suppressed, while expression of the corresponding human orthologs was unaffected. As androgen-deprived LNCaP cells up-regulate Shh expression, but are not sensitive to Smo antagonists, these studies indicate that TAK-441 leads to delayed castration-resistant progression of LNCaP xenografts by disrupting paracrine Hh signaling with the tumor stroma. Thus, paracrine Hh signaling may offer unique opportunities for prognostic biomarker development, drug targeting and therapeutic response monitoring of PCa progression.


Assuntos
Antineoplásicos/farmacologia , Comunicação Parácrina/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Piridinas/farmacologia , Pirróis/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Castração , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias da Próstata/metabolismo , Receptor Smoothened , Microambiente Tumoral/efeitos dos fármacos , Alcaloides de Veratrum/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Carcinog ; 51(12): 952-62, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22006862

RESUMO

Apigenin (4',5,7,-trihydroxyflavone), an anticancer agent, selectively toxic to cancer cells induces cell cycle arrest and apoptosis through mechanisms that have not been fully elucidated. Our studies indicate that apigenin-mediated growth inhibitory responses are due to inhibition of class I histone deacetylases (HDACs) in prostate cancer cells. Treatment of PC-3 and 22Rv1 cells with apigenin (20-40 µM) resulted in the inhibition of HDAC enzyme activity, specifically HDAC1 and HDAC3 at the protein and message level. Apigenin-mediated HDAC inhibition resulted in global histone H3 and H4 acetylation, as well as localized hyperacetylation of histone H3 on the p21/waf1 promoter. A corresponding increase was observed in p21/waf1 and bax protein and mRNA expression after apigenin exposure, consistent with the use of HDAC inhibitor, trichostatin A. The downstream events demonstrated cell cycle arrest and induction of apoptosis in both cancer cells. Studies of PC-3 xenografts in athymic nude mice further demonstrated that oral intake of apigenin at doses of 20 and 50 µg/mouse/d over an 8-wk period resulted in a marked reduction in tumor growth, HDAC activity, and HDAC1 and HDAC3 protein expression at both doses of apigenin. An increase in p21/waf1 expression was observed in apigenin-fed mice, compared to the control group. Furthermore, apigenin intake caused a significant decrease in bcl2 expression with concomitant increase in bax, shifting the bax/bcl2 ratio in favor of apoptosis. Our findings confirm for the first time that apigenin inhibits class I HDACs, particularly HDAC1 and HDAC3 and its exposure results in reversal of aberrant epigenetic events that promote malignancy. © 2011 Wiley Periodicals, Inc.


Assuntos
Apigenina/farmacologia , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Plantas/química , Neoplasias da Próstata/patologia , Acetilação , Animais , Sequência de Bases , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Inibidor de Quinase Dependente de Ciclina p21/genética , Primers do DNA , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas
6.
Drug Metab Dispos ; 40(5): 907-18, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22301272

RESUMO

The biological activities of vitamin D(3) are exerted through the dihydroxy metabolite of vitamin D(3) [1α,25(OH)(2)D(3)]. Hepatic biotransformation of 1α,25(OH)(2)D(3) by cytochrome P450 (P450) enzymes could be an important determinant of bioavailability in serum and tissues. In the present study, we investigated the comparative biotransformation of 1α,25(OH)(2)D(3) in mouse and human liver microsomes and determined the effects of commonly used drugs on the catabolism of 1α,25(OH)(2)D(3). Severe symptoms of vitamin D deficiency have historically been observed in patients who received dexamethasone. To compare the effects of clinically important glucocorticoids with hepatic biotransformation of 1α,25(OH)(2)D(3), adult male CD-1 mice were given intraperitoneal injections of either vehicle (50% ethanol), dexamethasone (80 mg/kg per day), or prednisone (80 mg/kg per day) for three consecutive days. Hydroxy metabolite formation pattern and the extent of substrate depletion were similar in mouse liver microsomes (MLM) from vehicle- or prednisone-treated mice, whereas treatment with dexamethasone led to the emergence of additional metabolites and increased substrate depletion, as determined by liquid chromatography/mass spectrometry. The metabolite formation profile in vehicle-treated mice was different from that of human liver microsomes (HLM). Selective P450 chemical inhibitors have demonstrated that CYP3A isoforms are responsible for the microsomal biotransformation of 1α,25(OH)(2)D(3) in MLM. Coincubation of 1α,25(OH)(2)D(3) with commonly used drugs led to approximately 60 to 100% inhibition of CYP3A4-mediated catabolism of 1α,25-(OH)(2)D(3) in HLM. A species-based difference was identified between CYP3A-mediated hepatic microsomal metabolism of 1α,25(OH)(2)D(3) in humans and mice. We have shown that the clinical importance of glucocorticoids differentially modulates catabolism of active vitamin D(3) and that commonly used drugs could affect vitamin D homeostasis.


Assuntos
Calcitriol/metabolismo , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450 , Inibidores Enzimáticos/farmacologia , Glucocorticoides/farmacologia , Microssomos Hepáticos/metabolismo , Animais , Calcitriol/farmacocinética , Citocromo P-450 CYP3A/biossíntese , Inibidores do Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/biossíntese , Dexametasona/farmacologia , Indução Enzimática , Feminino , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos , Microssomos Hepáticos/enzimologia , Prednisona/farmacologia , Especificidade da Espécie
7.
Pharmaceutics ; 13(9)2021 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-34575583

RESUMO

There have been several studies that have linked elevated scavenger receptor class b type 1 (SR-B1) expression and activity to the development and progression of castration-resistant prostate cancer (CRPC). SR-B1 facilitates the influx of cholesterol to the cell from lipoproteins in systemic circulation. This influx of cholesterol may be important for many cellular functions, including the synthesis of androgens. Castration-resistant prostate cancer tumors can synthesize androgens de novo to supplement the loss of exogenous sources often induced by androgen deprivation therapy. Silencing of SR-B1 may impact the ability of prostate cancer cells, particularly those of the castration-resistant state, to maintain the intracellular supply of androgens by removing a supply of cholesterol. SR-B1 expression is elevated in CRPC models and has been linked to poor survival of patients. The overarching belief has been that cholesterol modulation, through either synthesis or uptake inhibition, will impact essential signaling processes, impeding the proliferation of prostate cancer. The reduction in cellular cholesterol availability can impede prostate cancer proliferation through both decreased steroid synthesis and steroid-independent mechanisms, providing a potential therapeutic target for the treatment of prostate cancer. In this article, we discuss and highlight the work on SR-B1 as a potential novel drug target for CRPC management.

8.
Int J Cancer ; 126(11): 2520-33, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19856314

RESUMO

Epigenetic silencing of gluthathione-S-transferase pi (GSTP1) is recognized as being a molecular hallmark of human prostate cancer. We investigated the effects of green tea polyphenols (GTPs) on GSTP1 re-expression and further elucidated its mechanism of action and long-term safety, compared with nucleoside-analog inhibitor of DNA methyltransferase (DNMT), 5-aza-2'-deoxycitidine. Exposure of human prostate cancer LNCaP cells to 1-10 microg/ml of GTP for 1-7 days caused a concentration- and time-dependent re-expression of GSTP1, which correlated with DNMT1 inhibition. Methyl-specific-PCR and sequencing revealed extensive demethylation in the proximal GSTP1 promoter and regions distal to the transcription factor binding sites. GTP exposure in a time-dependent fashion diminished the mRNA and protein levels of MBD1, MBD4 and MeCP2; HDAC 1-3 and increased the levels of acetylated histone H3 (LysH9/18) and H4. Chromatin immunoprecipitation assays demonstrated that cells treated with GTP have reduced MBD2 association with accessible Sp1 binding sites leading to increased binding and transcriptional activation of the GSTP1 gene. Exposure of cells to GTP did not result in global hypomethylation, as demonstrated by methyl-specific PCR for LINE-1 promoter; rather GTP promotes maintenance of genomic integrity. Furthermore, exposure of cells to GTP did not cause activation of the prometaststic gene S100P, a reverse response noted after exposure of cells to 5-aza-2'deoxycitidine. Our results, for the first time, demonstrate that GTP has dual potential to alter DNA methylation and chromatin modeling, the 2 global epigenetic mechanisms of gene regulation and their lack of toxicity makes them excellent candidates for the chemoprevention of prostate cancer.


Assuntos
Flavonoides/farmacologia , Glutationa S-Transferase pi/genética , Fenóis/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/genética , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , Células Epiteliais/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Guanosina Trifosfato/farmacologia , Histonas/isolamento & purificação , Humanos , Imunoglobulinas/genética , Imuno-Histoquímica , Masculino , Polifenóis , Próstata/citologia , Próstata/fisiologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Chá/química
9.
Front Genet ; 10: 235, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30972102

RESUMO

Prostate cancer remains a major public health problem and the second leading cause of cancer-related deaths in men in the United States. The present study aims to understand the molecular pathway(s) of prostate cancer which is essential for early detection and treatment. Dorsolateral prostate from 20 week transgenic adenocarcinoma of the mouse prostate (TRAMP) mice, which spontaneously develops prostate cancer and recapitulates human disease and age-matched non-transgenic littermates were utilized for microarray analysis. Mouse genome network and pathway analyses were mapped to the human genome using the Ingenuity Pathway Analysis (IPA) database for annotation, visualization, and integrated discovery. In total, 136 differentially expressed genes, including 32 downregulated genes and 104 upregulated genes were identified in the dorsolateral prostate of TRAMP, compared to non-transgenic mice. A subset of differentially expressed genes were validated by qRT-PCR. Alignment with human genome database identified 18 different classes of proteins, among these, 36% were connected to the nucleic acid binding, including ribosomal proteins, which play important role in protein synthesis-the most enriched pathway in the development of prostate cancer. Furthermore, the results suggest deregulation of signaling molecules (9%) and enzyme modulators (8%) affect various pathways. An imbalance in other protein classes, including transporter proteins (7%), hydrolases (6%), oxidoreductases, and cytoskeleton proteins (5%), contribute to cancer progression. Our study evaluated the underlying pathways and its connection to human prostate cancer, which may further help assess the risk of disease development and progression and identify potential targets for therapeutic intervention.

10.
PLoS One ; 14(11): e0224071, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31756185

RESUMO

Dysregulation of miRNAs has been demonstrated in several human malignancies including prostate cancer. Due to tissue limitation and variable disease progression, stage-specific miRNAs changes in prostate cancer is unknown. Using chip-based microarray, we investigated global miRNA expression in human prostate cancer LNCaP, PC3, DU145 and 22Rv1 cells representing early-stage, advanced-stage and castration resistant prostate cancer in comparison with normal prostate epithelial cells. A total of 292 miRNAs were differentially expressed with 125 upregulated and 167 downregulated. These miRNAs were involved in pathways including drug resistance drug-efflux, adipogenesis, epithelial-to-mesenchymal transition, bone metamorphosis, and Th1/Th2 signaling. Regulation of miRNAs were interlinked with upstream regulators such as Argonaut 2 (AGO2), Double-Stranded RNA-Specific Endoribonuclease (DICER1), Sjogren syndrome antigen B (SSB), neurofibromatosis 2 (NF2), and peroxisome proliferator activated receptor alpha (PPARA), activated during stage-specific disease progression. Candidate target genes and pathways dysregulated in stage-specific prostate cancer were identified using CS-miRTar database and confirmed in clinical specimens. Integrative network analysis suggested some genes targeted by miRNAs include miR-17, let7g, miR-146, miR-204, miR-205, miR-221, miR-301 and miR-520 having a major effect on their dysregulation in prostate cancer. MiRNA-microarray analysis further identified miR-130a, miR-181, miR-328, miR146 and miR-200 as a panel of novel miRNAs associated with drug resistance drug-efflux and epithelial-to-mesenchymal transition in prostate cancer. Our findings provide evidence on miRNA dysregulation and its association with key functional components in stage-specific prostate cancer.


Assuntos
Biologia Computacional/métodos , Perfilação da Expressão Gênica , MicroRNAs/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Linhagem Celular Tumoral , Humanos , Masculino , Estadiamento de Neoplasias , Transcrição Gênica
11.
Methods Mol Med ; 116: 81-101, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16000856

RESUMO

Inhibition of protein synthesis by interferon treatment is mediated by two major pathways: the 2'-5'-linked oligoadenylates [2-5 (A)] synthetase-RNase L pathway and the double-stranded ribonucleic acid-dependent protein kinase-mediated pathway. 2-5 (A) synthetases are unique interferon-inducible enzymes that, upon activation by double-stranded RNA, polymerize adenosine triphosphate (ATP) to 2-5 (A) synthases. These 2-5 (A) synthetases bind and activate the latent RNase L, causing RNA degradation. In addition to the three major size classes of enzymatically active oligoadenylate synthetase proteins, at least one inactive oligoadenylate synthetase is known in human and mouse. Structure-function studies and recent crystal structure determination have identified several distinct sites in these proteins responsible for different biochemical functions. RNase L is the only known protein that binds to 2-5 (A) synthetases with very high affinity. Gene knockout studies of RNase L have identified its role in antiviral actions of interferon and in apoptosis. Recently, it has also been implicated in prostate cancer metastasis. In this chapter we describe several methodologies for studying biochemical and physiological properties of the 2-5 (A) synthetase-RNase L pathway.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , Bioensaio/métodos , Interferons/metabolismo , Isoenzimas/metabolismo , 2',5'-Oligoadenilato Sintetase/genética , Animais , Reagentes de Ligações Cruzadas/metabolismo , Endorribonucleases/genética , Endorribonucleases/metabolismo , Humanos , Isoenzimas/genética , Camundongos , Ligação Proteica , RNA/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
12.
RNA Biol ; 1(1): 21-7, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-17200614

RESUMO

RNA-degradation is one of the fundamental mechanisms of interferon (IFN)-inducible antiviral response in mammalian cells. This is primarily brought about by the IFN-inducible 2',5'-oligoadenylate (2-5A)-cofactor dependent ribonuclease L (RNase L). RNase L also functions as a tumor suppressor gene in case of prostate cancer due to its role in apoptosis. We report that RNase L is induced by stress-inducing agents such as double-stranded RNA [poly(I:C)], chemotherapeutic drugs, hydrogen peroxide (H(2)O(2)), calcium chloride (CaCl(2)) and tumor necrosis factor-alpha (TNF) in the human cervical carcinoma (HeLa) cells. The level of RNase L was not detected in the untreated cells. Induction of RNase L by such stress-inducing agents correlated with degradation of cellular RNA, fragmentation of chromatin-DNA and induction of apoptosis. We checked the stress-inducible transcription factor, nuclear factor kappa B (NF-kappaB), which was persistently activated by cycloheximide but not by other agents after 24 hours indicating no role of NFkappaB in the RNase L-induction. However, as expected, TNF-induced NF-kappaB activity was stimulated within 10-30 minutes through degradation of IkappaB-alpha. Our results strongly suggest that the IFN-inducible RNase L is induced by a broad range of stress-inducing signals such as double-stranded RNA (dsRNA) produced during viral infection, membrane- and osmotic shock caused by CaCl(2) and oxidative stress induced by H(2)O(2), inflammation stimulated by TNF-alpha and chemotherapy. Thus, in addition to its antiviral function, the IFN-inducible RNase L may play an important role during stress-response through RNA-degradation and apoptosis.


Assuntos
Carcinoma/enzimologia , Endorribonucleases/metabolismo , Neoplasias do Colo do Útero/enzimologia , Antineoplásicos/farmacologia , Apoptose , Cloreto de Cálcio/farmacologia , Feminino , Humanos , Interferons/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo , Poli I-C/farmacologia , RNA/química , RNA de Cadeia Dupla/farmacologia , Fatores de Tempo
13.
Mol Cancer Ther ; 13(12): 2827-39, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25267499

RESUMO

Insulin-like growth factor (IGF) signaling is associated with castrate-resistant prostate cancer (CRPC) progression. Insulin receptor substrates 1 and 2 (IRS1/2) mediate mitogenic and antiapoptotic signaling from IGF1 receptor (IGF1R), insulin receptor, and other oncoproteins. This study demonstrates that IRS1/2 expression is increased in prostate cancer, and persists in CRPC. Furthermore, this study assesses the anticancer activity of NT157, a small molecule tyrphostin targeting IRS proteins, using androgen-responsive (LNCaP) and -independent (PC3) prostate cancer cells in vitro and in vivo. NT157 treatment resulted in dose-dependent inhibition of IGF1R activation, suppression of IRS protein expression, inhibition of IGF1-induced AKT activation, but increased ERK activation in NT157-treated cells in vitro. These effects were correlated with decreased proliferation and increasing apoptosis of LNCaP cells and increasing G2-M arrest in PC3 cells. NT157 also suppressed androgen-responsive growth, delayed CRPC progression of LNCaP xenografts, and suppressed PC3 tumor growth alone and in combination with docetaxel. This study reports the first preclinical proof-of-principle data that this novel small molecule tyrosine kinase inhibitor suppresses IRS1/2 expression, delays CRPC progression, and suppresses growth of CRPC tumors in vitro and in vivo. Demonstration that IRS expression can be increased in response to a variety of stressors that may lead to resistance or reduced effect of the therapies indicate that NT157-mediated IRS1/2 downregulation is a novel therapeutic approach for management of advanced prostate cancer.


Assuntos
Proteínas Substratos do Receptor de Insulina/metabolismo , Neoplasias da Próstata/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Tirfostinas/farmacologia , Animais , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Docetaxel , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Substratos do Receptor de Insulina/genética , Masculino , Camundongos , Orquiectomia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteólise , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Taxoides/farmacologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Front Biosci (Elite Ed) ; 1(1): 13-25, 2009 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-19482620

RESUMO

Green tea, the most popular beverage next to water, is a rich source of tea catechins and has potential to be developed as a chemopreventive agent for prostate cancer. For centuries it has been used in traditional medicine in Far-East countries. Male populations in these countries where large quantities of green tea are consumed on regular basis have the lowest incidence of prostate cancer. In this review, after a description of prostate cancer and several risk factors associated with the disease, we evaluated studies reported with green tea or its major constituent, (-)-epigallocatechin-3-gallate in inhibition of prostate cancer. This review provides an in-depth overview of various biochemical and signaling pathways affected by green tea in in vivo and in vitro models of prostate cancer. This is followed by a comprehensive discussion of the epidemiological studies and some ongoing clinical trials with green tea catechins. The review concludes with a brief discussion of the future direction and development of clinical trials employing green tea catechins which could be developed for prevention and/or intervention of prostate cancer.


Assuntos
Anticarcinógenos/farmacologia , Camellia sinensis/química , Catequina/análogos & derivados , Fitoterapia/métodos , Extratos Vegetais/farmacologia , Neoplasias da Próstata/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Animais , Catequina/farmacologia , Humanos , Masculino , Camundongos , Neoplasias da Próstata/epidemiologia
15.
Life Sci ; 85(19-20): 663-9, 2009 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-19788894

RESUMO

AIMS: Inducible cyclooxygenase (COX-2) has been implicated in the process of inflammation and carcinogenesis. Chamomile has long been used in traditional medicine for the treatment of inflammatory diseases. In this study we aimed to investigate whether chamomile interferes with the COX-2 pathway. MAIN METHODS: We used lipopolysaccharide (LPS)-activated RAW 264.7 macrophages as an in vitro model for our studies. KEY FINDINGS: Chamomile treatment inhibited the release of LPS-induced prostaglandin E(2) in RAW 264.7 macrophages. This effect was found to be due to inhibition of COX-2 enzyme activity by chamomile. In addition, chamomile caused reduction in LPS-induced COX-2 mRNA and protein expression, without affecting COX-1 expression. The non-steroidal anti-inflammatory drug, sulindac and a specific COX-2 inhibitor, NS398, were shown to act similarly in LPS-activated RAW 264.7 cells. Our data suggest that chamomile works by a mechanism of action similar to that attributed to non-steroidal anti-inflammatory drugs. SIGNIFICANCE: These findings add a novel aspect to the biological profile of chamomile which might be important for understanding the usefulness of aqueous chamomile extract in the form of tea in preventing inflammation and cancer.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Camomila/química , Inibidores de Ciclo-Oxigenase 2/farmacologia , Animais , Apigenina/química , Apigenina/farmacologia , Western Blotting , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/genética , Dinoprostona/biossíntese , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Camundongos , Extratos Vegetais/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Mol Biol Rep ; 34(2): 97-104, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17091337

RESUMO

Interferons (IFNs) induce a 2',5'-oligoadenylate (2-5A)-dependent ribonuclease L (RNase L) following virus-infection of mammalian cells. RNase L degrades both viral and cellular RNAs and restricts virus-proliferation. We have studied organization of RNase L gene in genomic DNA from the mouse liver by Southern blot analysis. Several BamHI, BglII, EcoRI, HincII, HindIII, NcoI, PstI, SacI, and XbaI restriction fragments hybridized to (32)P-labeled mouse RNase L cDNA and the 5'-proximal exon probes. Mouse RNase L gene exists as a single copy (>16 kb DNA) gene. A 5 kb HindIII and a 2.5 kb EcoRI DNA were detected as 5'-upstream DNA of the gene which may contain mouse RNase L promoter. Our results will help studying mouse RNase L gene promoter in further details.


Assuntos
Endorribonucleases/genética , Região 5'-Flanqueadora , Animais , Endorribonucleases/química , Endorribonucleases/metabolismo , Éxons , Feminino , Interferons/fisiologia , Camundongos , Mapeamento por Restrição , Transdução de Sinais
17.
Biochemistry ; 44(18): 6837-43, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15865429

RESUMO

Unlike other RNA polymerases, 2'-5' oligoadenylate synthetases, a family of interferon-induced enzymes, catalyze the formation of 2'-5', not 3'-5', phosphodiester bonds. Moreover, to be active, these proteins require double-stranded RNA as a cofactor. We have been identifying the specific residues of these proteins that impart their novel properties. Here, we report the identity of three such residues that underwent natural mutations in a transgenic mouse line. When deliberately introduced into recombinant proteins, each of these mutations rendered the protein enzymatically inactive. In an effort to understand the roles of these residues in enzyme activity, new mutants carrying other residues in one of these three sites were generated. Detailed characterization of the properties of the mutant proteins revealed that Lys 404 is needed for proper binding of the acceptor substrate, Pro 500 provides structural flexibility to the protein, and Ser 471 is probably required for its proper folding. This study illustrates the power of using natural mutations in transgenes as guides for studying structure-function relationships of proteins.


Assuntos
2',5'-Oligoadenilato Sintetase/química , 2',5'-Oligoadenilato Sintetase/genética , Lisina/genética , Prolina/genética , Serina/genética , Transgenes , Sequência de Aminoácidos , Animais , Ativação Enzimática/genética , Glicina/genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida
18.
Biochem Biophys Res Commun ; 317(2): 586-97, 2004 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-15063798

RESUMO

Interferon-inducible ribonuclease L (RNase L) is a unique ankyrin-repeat containing endoribonuclease activated by 2',5'-oligoadenylate (2-5A) cofactor leading to RNA degradation and apoptosis during antiviral response in mammalian cells. We report that expression of recombinant human RNase L (1-741 a.a.) caused RNA degradation and inhibition of cell growth in Escherichia coli in absence of exogenous 2-5A. On the contrary, expression of a homologous but dominant negative form of murine RNase L (1-656 a.a.), lacking the RNA binding and ribonuclease domain, did not show RNA degradation, rather it stimulated cell growth. Upon computational analysis by pBLAST search, a putative transcription factor (yahD, F64758, and NP_414852) from the E. coli genome showed highest homology (E value=1e(-17)) with 90-259 a.a. region of human RNase L due to ankyrin repeats with conserved GKT motifs. Ankyrin repeats 6-9 of RNase L are involved in 2-5A binding, dimerization, and activation of the ribonuclease. Thus, a biochemically active human RNase L in E. coli strongly suggests for a prokaryotic cell growth-inhibitory mechanism possibly through ankyrin-ankyrin interaction of YahD and RNase L leading to RNA degradation. The mammalian interferon-inducible RNase L and E. coli yahD protein may have common origin for the ankyrin repeats with 2-5A binding sites. Thus, RNA degradation and cell growth inhibition by recombinant human RNase L biochemically reconstituted mammalian cellular response to interferon in E. coli. RNase L has prokaryotic evolutionary history, it is not only an antiviral but also an antibacterial gene.


Assuntos
Proteínas de Transporte/metabolismo , Endorribonucleases/química , Endorribonucleases/metabolismo , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , RNA/metabolismo , Sequência de Aminoácidos , Divisão Celular/fisiologia , Coenzimas/metabolismo , Endorribonucleases/genética , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Inibidores do Crescimento/genética , Inibidores do Crescimento/metabolismo , Humanos , Dados de Sequência Molecular , RNA/química , Proteínas Recombinantes/biossíntese , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa