Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Int J Mol Sci ; 22(2)2021 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-33418978

RESUMO

The peripheral zone (PZ) and transition zone (TZ) represent about 70% of the human prostate gland with each zone having differential ability to develop prostate cancer. Androgens and their receptor are the primary driving cause of prostate cancer growth and eventually castration-resistant prostate cancer (CRPC). De novo steroidogenesis has been identified as a key mechanism that develops during CRPC. Currently, there is very limited information available on human prostate tissue steroidogenesis. The purpose of the present study was to investigate steroid metabolism in human prostate cancer tissues with comparison between PZ and TZ. Human prostate cancer tumors were procured from the patients who underwent radical prostatectomy without any neoadjuvant therapy. Human prostate homogenates were used to quantify steroid levels intrinsically present in the tissues as well as formed after incubation with 2 µg/mL of 17-hydroxypregnenolone (17-OH-pregnenolone) or progesterone. A Waters Acquity ultraperformance liquid chromatography coupled to a Quattro Premier XE tandem quadrupole mass spectrometer using a C18 column was used to measure thirteen steroids from the classical and backdoor steroidogenesis pathways. The intrinsic prostate tissue steroid levels were similar between PZ and TZ with dehydroepiandrosterone (DHEA), dihydrotestosterone (DHT), pregnenolone and 17-OH-pregnenolone levels higher than the other steroids measured. Interestingly, 5-pregnan-3,20-dione, 5-pregnan-3-ol-20-one, and 5-pregnan-17-ol-3,20-dione formation was significantly higher in both the zones of prostate tissues, whereas, androstenedione, testosterone, DHT, and progesterone levels were significantly lower after 60 min incubation compared to the 0 min control incubations. The incubations with progesterone had a similar outcome with 5-pregnan-3,20-dione and 5-pregnan-3-ol-20-one levels were elevated and the levels of DHT were lower in both PZ and TZ tissues. The net changes in steroid formation after the incubation were more observable with 17-OH-pregnenolone than with progesterone. In our knowledge, this is the first report of comprehensive analyses of intrinsic prostate tissue steroids and precursor-driven steroid metabolism using a sensitive liquid chromatography-mass spectrometry assay. In summary, the PZ and TZ of human prostate exhibited similar steroidogenic ability with distinction in the manner each zone utilizes the steroid precursors to divert the activity towards backdoor pathway through a complex matrix of steroidogenic mechanisms.


Assuntos
Neoplasias da Próstata/patologia , Esteroides/metabolismo , Androstenodiona/análise , Androsterona/análise , Cromatografia Líquida de Alta Pressão , Humanos , Masculino , Espectrometria de Massas , Progesterona/análogos & derivados , Progesterona/análise , Progesterona/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Esteroides/análise , Esteroides/química , Testosterona/análise
2.
Medicines (Basel) ; 8(6)2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-34199743

RESUMO

In spite of possessing desirable anticancer properties, currently, limited clinical success has been achieved with 20(S)-protopanaxadiol (aPPD) and 1,25-dihydroxyvitamin D3 (calcitriol). This study is designed to evaluate if the combination of aPPD with calcitriol can inhibit human prostate cancer xenograft growth by using nuclear receptor signaling. Athymic male nude mice were utilized to establish an androgen-independent human prostate cancer C4-2 cell castration-resistant prostate cancer (CRPC) xenograft model. Mice were treated orally for six weeks with 70 mg/kg aPPD administered once daily or three times per week with 4 µg/kg calcitriol or in combination or only with vehicle control. Contrary to our expectations, calcitriol treatment alone increased C4-2 tumor growth. However, the addition of calcitriol substantially increased aPPD-mediated tumor growth suppression (76% vs. 53%, combination vs. aPPD alone). The combination treatment significantly increased levels of cleaved caspase-3 apoptotic marker compared to vehicle-treated or aPPD-treated C4-2 tumors. The mechanistic elucidations indicate that tumor inhibition by the aPPD and calcitriol combination was accompanied by elevated vitamin D receptor (VDR) protein expression. In silico data suggest that aPPD weakly binds to the native LBD pocket of VDR. Interestingly, the combination of aPPD and calcitriol activated VDR at a significantly higher level than calcitriol alone and this indicates that aPPD may be an allosteric activator of VDR. Overall, aPPD and calcitriol combination significantly inhibited tumor growth in vivo with no acute or chronic toxic effects in the C4-2 xenograft CRPC nude mice. The involvement of VDR and downstream apoptotic pathways are potential mechanistic routes of antitumor effects of this combination.

3.
Cancer Res ; 66(4): 1906-11, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16488987

RESUMO

p33ING2 is a novel candidate tumor suppressor, which has been shown to be involved in the regulation of gene transcription, cell cycle arrest, and apoptosis in a p53-dependent manner for maintaining the genomic stability. Previously, we showed that p33ING2 promoted UV-induced apoptosis in human melanoma cells. To further reveal the role of p33ING2 in cellular stress response to UV irradiation, we hypothesized that p33ING2 may enhance the repair of UV-damaged DNA, similarly to its homologue p33(ING1b). Using the host-cell reactivation assay, we show that overexpression of p33ING2 significantly enhances nucleotide excision repair of UV-induced DNA damage in melanoma cells in a p53-dependent manner. Furthermore, DNA repair is completely abolished in cells treated with p33ING2 small interfering RNA, suggesting that a physiologic level of p33ING2 is required for nucleotide excision repair. In addition, we found that p33ING2 is an essential factor for UV-induced rapid histone H4 acetylation, chromatin relaxation, and the recruitment of damage recognition protein, xeroderma pigmentosum group A protein, to the photolesions. These observations suggest that p33ING2 is required for the initial DNA damage sensing and chromatin remodeling in the nucleotide excision repair process.


Assuntos
Cromatina/metabolismo , Reparo do DNA/fisiologia , Histonas/metabolismo , Proteínas de Homeodomínio/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Acetilação/efeitos da radiação , Dano ao DNA , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Melanoma/genética , Melanoma/metabolismo , RNA Interferente Pequeno/genética , Receptores Citoplasmáticos e Nucleares , Transfecção , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Raios Ultravioleta , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo
4.
Cancers (Basel) ; 10(10)2018 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-30241348

RESUMO

Castration-resistant prostate tumors acquire the independent capacity to generate androgens by upregulating steroidogenic enzymes or using steroid precursors produced by the adrenal glands for continued growth and sustainability. The formation of steroids was measured by liquid chromatography-mass spectrometry in LNCaP and 22Rv1 prostate cancer cells, and in human prostate tissues, following incubation with steroid precursors (22-OH-cholesterol, pregnenolone, 17-OH-pregnenolone, progesterone, 17-OH-progesterone). Pregnenolone, progesterone, 17-OH-pregnenolone, and 17-OH-progesterone increased C21 steroid (5-pregnan-3,20-dione, 5-pregnan-3,17-diol-20-one, 5-pregnan-3-ol-20-one) formation in the backdoor pathway, and demonstrated a trend of stimulating dihydroepiandrosterone or its precursors in the backdoor pathway in LNCaP and 22Rv1 cells. The precursors differentially affected steroidogenic enzyme messenger RNA (mRNA) expressions in the cell lines. The steroidogenesis following incubation of human prostate tissue with 17-OH-pregnenolone and progesterone produced trends similar to those observed in cell lines. Interestingly, the formation of C21 steroids from classical pathway was not stimulated but backdoor pathway steroids (e.g., 5-pregnan-3,20-dione, 5-pregnan-3-ol-20-one) were elevated following incubations with prostate tissues. Overall, C21 steroids were predominantly formed in the classical as well as backdoor pathways, and steroid precursors induced a diversion of steroidogenesis to the backdoor pathway in both cell lines and human prostate tissue, and influenced adaptive steroidogenesis to form C21 steroids.

5.
Oncotarget ; 9(30): 20965-20978, 2018 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-29765513

RESUMO

We have explored the effects of 20(S)-protopanaxadiol (aPPD), a naturally derived ginsenoside, against androgen receptor (AR) positive castration resistant prostate cancer (CRPC) xenograft tumors and have examined its interactions with AR. In silico docking studies for aPPD binding to AR, alongside transactivation bioassays and in vivo efficacy studies were carried out in the castration-resistant C4-2 xenograft model. Immunohistochemical (IHC) and Western blot analyses followed by evaluation of AR, apoptotic, cell cycle and proliferative markers in excised tumors was performed. The growth of established CRPC tumors was inhibited by 53% with aPPD and a corresponding decrease in serum PSA was seen compared to controls. The IHC data revealed that Ki-67 was significantly lower for aPPD treated tumors and was associated with elevated p21 and cleaved caspase-3 expression, compared to vehicle treatment. Furthermore, aPPD decreased AR protein expression in xenograft tumors, while significantly upregulating p27 and Bax protein levels. In vitro data supporting this suggests that aPPD binds to and significantly inhibits the N-terminal or the DNA binding domains of AR. The AR androgen binding site docking score for androgen (dihydrotestosterone) was -11.1, while that of aPPD was -7.1. The novel findings described herein indicate aPPD potently inhibits PCa in vivo partly via inhibition of a site on the AR N-terminal domain. This manifested as cell cycle arrest and concurrent induction of apoptosis via an increase in Bax, cleaved-caspase-3, p27 and p21 expression.

6.
Sci Rep ; 8(1): 2090, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29391407

RESUMO

The molecular chaperone Hsp90 is overexpressed in prostate cancer (PCa) and is responsible for the folding, stabilization and maturation of multiple oncoproteins, which are implicated in PCa progression. Compared to first-in-class Hsp90 inhibitors such as 17-allylamino-demethoxygeldanamycin (17-AAG) that were clinically ineffective, second generation inhibitor AUY922 has greater solubility and efficacy. Here, transcriptomic and proteomic analyses of patient-derived PCa explants identified cytoskeletal organization as highly enriched with AUY922 treatment. Validation in PCa cell lines revealed that AUY922 caused marked alterations to cell morphology, and suppressed cell motility and invasion compared to vehicle or 17-AAG, concomitant with dysregulation of key extracellular matrix proteins such as fibronectin (FN1). Interestingly, while the expression of FN1 was increased by AUY922, FN1 secretion was significantly decreased. This resulted in cytosolic accumulation of FN1 protein within late endosomes, suggesting that AUY922 disrupts vesicular secretory trafficking pathways. Depletion of FN1 by siRNA knockdown markedly reduced the invasive capacity of PCa cells, phenocopying AUY922. These results highlight a novel mechanism of action for AUY922 beyond its established effects on cellular mitosis and survival and, furthermore, identifies extracellular matrix cargo delivery as a potential therapeutic target for the treatment of aggressive PCa.


Assuntos
Fibronectinas/metabolismo , Neoplasias da Próstata/metabolismo , Via Secretória/efeitos dos fármacos , Idoso , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Fibronectinas/genética , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Isoxazóis/farmacologia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Neoplasias da Próstata/patologia , Resorcinóis/farmacologia
7.
PLoS One ; 11(5): e0154967, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27152724

RESUMO

Exosomes proteins and microRNAs have gained much attention as diagnostic tools and biomarker potential in various malignancies including prostate cancer (PCa). However, the role of exosomes and membrane-associated receptors, particularly epidermal growth factor receptor (EGFR) as mediators of cell proliferation and invasion in PCa progression remains unexplored. EGFR is frequently overexpressed and has been associated with aggressive forms of PCa. While PCa cells and tissues express EGFR, it is unknown whether exosomes derived from PCa cells or PCa patient serum contains EGFR. The aim of this study was to detect and characterize EGFR in exosomes derived from PCa cells, LNCaP xenograft and PCa patient serum. Exosomes were isolated from conditioned media of different PCa cell lines; LNCaP xenograft serum as well as patient plasma/serum by differential centrifugation and ultracentrifugation on a sucrose density gradient. Exosomes were confirmed by electron microscopy, expression of exosomal markers and NanoSight™ analysis. EGFR expression was determined by western blot analysis and ELISA. This study demonstrates that exosomes may easily be derived from PCa cell lines, serum obtained from PCa xenograft bearing mice and clinical samples derived from PCa patients. Presence of exosomal EGFR in PCa patient exosomes may present a novel approach for measuring of the disease state. Our work will allow to build on this finding for future understanding of PCa exosomes and their potential role in PCa progression and as minimal invasive biomarkers for PCa.


Assuntos
Receptores ErbB/metabolismo , Exossomos/metabolismo , Neoplasias da Próstata/metabolismo , Animais , Biomarcadores Tumorais/sangue , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Xenoenxertos , Humanos , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia
8.
PLoS One ; 11(6): e0157392, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27295211

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0154967.].

9.
Oncotarget ; 7(12): 14639-58, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26840259

RESUMO

Prostate cancer (PCa) is the most frequently diagnosed cancer in men. Current research on tumour-related extracellular vesicles (EVs) suggests that exosomes play a significant role in paracrine signaling pathways, thus potentially influencing cancer progression via multiple mechanisms. In fact, during the last decade numerous studies have revealed the role of EVs in the progression of various pathological conditions including cancer. Moreover, differences in the proteomic, lipidomic, and cholesterol content of exosomes derived from PCa cell lines versus benign prostate cell lines confirm that exosomes could be excellent biomarker candidates. As such, as part of an extensive proteomic analysis using LCMS we previously described a potential role of exosomes as biomarkers for PCa. Current evidence suggests that uptake of EV's into the local tumour microenvironment encouraging us to further examine the role of these vesicles in distinct mechanisms involved in the progression of PCa and castration resistant PCa. For the purpose of this study, we hypothesized that exosomes play a pivotal role in cell-cell communication in the local tumour microenvironment, conferring activation of numerous survival mechanisms during PCa progression and development of therapeutic resistance. Our in vitro results demonstrate that PCa derived exosomes significantly reduce apoptosis, increase cancer cell proliferation and induce cell migration in LNCaP and RWPE-1 cells. In conjunction with our in vitro findings, we have also demonstrated that exosomes increased tumor volume and serum PSA levels in vivo when xenograft bearing mice were administered DU145 cell derived exosomes intravenously. This research suggests that, regardless of androgen receptor phenotype, exosomes derived from PCa cells significantly enhance multiple mechanisms that contribute to PCa progression.


Assuntos
Comunicação Celular , Proliferação de Células , Exossomos/patologia , Neoplasias da Próstata/patologia , Microambiente Tumoral , Animais , Apoptose , Movimento Celular , Progressão da Doença , Exossomos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Fenótipo , Neoplasias da Próstata/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Steroid Biochem Mol Biol ; 144 Pt A: 50-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24269662

RESUMO

The chemopreventive and therapeutic effects of vitamin D3 are exerted through its dihydroxylated metabolite, 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3]. Inactivation of 1α,25(OH)2D3 by cytochrome P450 3A4 (CYP3A4) may be an important determinant of its serum and tissue levels. Abiraterone, a steroidogenesis inhibitor used in late stage prostate cancer treatment, is a CYP17A1 inhibitor. The purpose of this study was to assess the potential of abiraterone to block hepatic and intestinal inactivation of biologically active vitamin D3in vitro and to evaluate if abiraterone can alter CYP3A4 marker substrate activities. Biotransformation reactions were initiated with NADPH regenerating solutions following initial preincubation of pooled human hepatic or intestinal microsomal protein or human recombinant CYP3A4 supersomes with 1α,25(OH)2D3, midazolam or triazolam for 10min at 37°C. Formation of hydroxylated metabolites of 1α,25(OH)2D3, midazolam or triazolam was analyzed by liquid chromatography-mass spectrometry method. Co-incubation of 1α,25(OH)2D3 with abiraterone at varying concentrations (0.2-100µM) led to up to ∼85% inhibition of formation of hydroxylated metabolites of 1α,25(OH)2D3 thus preventing inactivation of active vitamin D3. The IC50 values for individual metabolites of 1α,25(OH)2D3 ranged from 0.4 to 2.2µM in human liver microsomes or human intestinal microsomes. The mechanism of CYP3A4-mediated inhibition of 1α,25(OH)2D3 by abiraterone was competitive (apparent Ki 2.8-4.3µM). Similar inhibitory effects were also observed upon inclusion of abiraterone into midazolam or triazolam hydroxylation assays. In summary, our results suggest that abiraterone inhibits the CYP3A4-mediated inactivation of active vitamin D3 in human liver and intestine, potentially providing additional anti-cancer benefits to prostate cancer patients. This article is part of a Special Issue entitled '16th Vitamin D Workshop'.


Assuntos
Androstenóis/farmacologia , Inibidores do Citocromo P-450 CYP3A , Citocromo P-450 CYP3A/metabolismo , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Vitamina D/análogos & derivados , Androstenos , Humanos , Técnicas In Vitro , Intestinos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Vitamina D/metabolismo
11.
J Steroid Biochem Mol Biol ; 141: 94-103, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24486455

RESUMO

The beneficial effects of vitamin D3 are exerted through 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3], the dihydroxy metabolite of vitamin D3. Hepatic and intestinal biotransformation of 1α,25(OH)2D3 and modifiers of metabolic capacity could be important determinants of bioavailability in serum and tissues. Ginsenosides and their aglycones, mainly 20(S)-protopanaxadiol (aPPD) and 20(S)-protopanaxatriol (aPPT), are routinely ingested as health supplements. The purpose of the present study was to investigate the potential of ginsenosides and their aglycones to block hepatic and intestinal inactivation of 1α,25(OH)2D3, which is the most potent ligand of vitamin D receptor. In vitro biotransformation reactions were initiated with NADPH regenerating solutions following initial preincubation of pooled human hepatic or intestinal microsomal protein or human recombinant CYP3A4 supersomes with 1α,25(OH)2D3 or midazolam. Formation of hydroxylated metabolites of 1α,25(OH)2D3 or midazolam was analyzed using liquid chromatography-mass spectrometry. Co-incubation of 1α,25(OH)2D3 with various ginsenosides (Rg1, Rh2, aPPD, aPPT and total ginsenosides) led to differential inhibition (30-100%) of its hydroxylation. Results suggest that aPPD, aPPT and Rh2 strongly attenuated the hydroxylation of 1α,25(OH)2D3. Follow up inhibition studies with aPPD and aPPT at varying concentrations (0.5-100µM) led to up to 91-100% inhibition of formation of hydroxylated metabolites of 1α,25(OH)2D3 thus preventing inactivation of active vitamin D3. The IC50 values of aPPD or aPPT for the most abundant hydroxylated metabolites of 1α,25(OH)2D3 ranged from 3.3 to 9.0µM in human microsomes. The inhibitory mechanism of aPPD or aPPT for CYP3A4-mediated biotransformation of 1α,25(OH)2D3 was competitive in nature (apparent Ki: 1.7-2.9µM). Similar inhibitory effects were also observed upon addition of aPPD or aPPT into midazolam hydroxylation assay. In summary, our results suggest that ginsenosides, specifically aPPD and aPPT, inhibit the CYP3A4-mediated catabolism of active vitamin D3 in human liver and intestine, potentially providing additional vitamin D-related benefits to patients with cancer, neurodegenerative and metabolic diseases.


Assuntos
Calcitriol/metabolismo , Sapogeninas/farmacologia , Biotransformação , Citocromo P-450 CYP3A/metabolismo , Inibidores do Citocromo P-450 CYP3A , Humanos , Intestinos/efeitos dos fármacos , Intestinos/enzimologia , Cinética , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Midazolam/farmacologia
12.
J Steroid Biochem Mol Biol ; 143: 19-28, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24565566

RESUMO

Castration-resistant prostate cancer (CRPC) remains largely dependent on androgen receptor (AR). Residual tissue androgens are consistently detected within CRPC tumors and play a critical role in facilitating AR-mediated signaling pathways which lead to disease progression. Testosterone and dihydrotestosterone (DHT) are the major androgens detected in tumors. They are produced through three biosynthesis pathways: Δ(4), Δ(5), and backdoor pathways. Both androgens bind to and stimulate AR activation. The current study investigates the effects of pomegranate extracts (POM) and their ability to inhibit androgen biosynthesis using PCa cell lines (22RV1 and LNCaP) in vitro as well as the PTEN knockout mouse model representing prostate cancer. Steroids were extracted using ethyl acetate or solid phase extraction, and then analyzed by UPLC/MS/MS. The results showed that POM (0-12µg/mL) reduced the production of testosterone, DHT, DHEA, androstenedione, androsterone, and pregnenolone in both cell lines. In addition our in vivo data supports this observation with a reduction in serum steroids determined after 20 weeks of POM treatment (0.17 g/L in drinking water). In accordance with these results, Western blotting of cell lysates and tPSA analysis determined that PSA was significantly decreased by the treatment of POM. Interestingly, AKR1C3 and AR levels were shown to be increased in both cell lines, perhaps as a negative feedback effect in response to steroid inhibition. Overall, these results provide mechanistic evidence to support the rationale for recent clinical reports describing efficacy of POM in CRPC patients.


Assuntos
Androgênios/biossíntese , Lythraceae/química , PTEN Fosfo-Hidrolase/fisiologia , Extratos Vegetais/farmacologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Androstenodiona/metabolismo , Animais , Western Blotting , Proliferação de Células/efeitos dos fármacos , Di-Hidrotestosterona/metabolismo , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Knockout , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/metabolismo , Testosterona/metabolismo , Células Tumorais Cultivadas
13.
Exp Cell Res ; 304(2): 531-43, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15748897

RESUMO

The roles of p33ING2 as a tumor suppressor candidate have been shown through regulation of gene transcription, induction of cell cycle arrest, and apoptosis. As p33ING2 shares 58.9% homology with p33ING1b, we hypothesized that p33ING2 shares functional similarities with p33ING1b. We previously found that p33ING1b cooperates with p53 to enhance UVB-induced apoptosis. Here, we report that overexpression of p33ING2 enhanced apoptosis in UVB-irradiated and non-irradiated melanoma MMRU cells. We demonstrate that enhancement of apoptosis by p33ING2 requires the presence of functional p53. Furthermore, we found that overexpression of p33ING2 significantly downregulated the expression of Bcl-2 after UVB irradiation, resulting in an increased Bax/Bcl-2 ratio. Moreover, we found that p33ING2 promoted Bax translocation to mitochondria, altered the mitochondrial membrane potential, and induced cytochrome c release and thus the activation of caspases 9 and 3. In addition, we showed that under non-stress conditions p33ING2 upregulates Fas expression and activates caspase 8. Taken together, we demonstrate that p33ING2 cooperates with p53 to regulate apoptosis via activation of both the mitochondrial/intrinsic and death-receptor/extrinsic apoptotic pathways.


Assuntos
Apoptose/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Proteínas de Homeodomínio/metabolismo , Melanoma/tratamento farmacológico , Proteínas Supressoras de Tumor/metabolismo , Raios Ultravioleta , Regulação para Cima/fisiologia , Apoptose/efeitos da radiação , Caspases/metabolismo , Citocromos c/metabolismo , Regulação para Baixo/fisiologia , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Proteínas de Homeodomínio/genética , Humanos , Melanoma/metabolismo , Melanoma/patologia , Potenciais da Membrana/fisiologia , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores Citoplasmáticos e Nucleares , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Regulação para Cima/efeitos da radiação , Proteína X Associada a bcl-2 , Receptor fas/metabolismo
14.
Exp Mol Pathol ; 75(2): 131-6, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14516774

RESUMO

Chk2 (Checkpoint kinase 2) is emerging as a critical mediator of genotoxic stress and diverse cellular responses. Upon ionizing radiation, Chk2 is activated to phosphorylate Cdc25C, leading to G2 phase arrest. p53 has been reported as another substrate of Chk2. Chk2 phosphorylates and stabilizes p53 in response to ionizing radiation. Previous studies found that p53 regulates the Chk2 homologue Chk1 expression both in vitro and in vivo. Using the p53-deficient mouse model, here we demonstrate by immunohistochemistry, Western blot analysis, and RT-PCR that mChk2 expression is reduced in the heart, kidney, lung, and liver of p53(-/-) mice compared to p53(+/+) controls. Similar Chk2 expression was observed in the brain, skin, spleen, and testis in p53(+/+) and p53(-/-) mice. These data indicate that p53 regulates Chk2 expression in a tissue-specific manner.


Assuntos
Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteína Supressora de Tumor p53/fisiologia , Animais , Western Blotting , Encéfalo/metabolismo , Quinase do Ponto de Checagem 2 , Regulação Enzimológica da Expressão Gênica/fisiologia , Genes Supressores de Tumor , Coração/fisiologia , Técnicas Imunoenzimáticas , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos , Proteínas Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/metabolismo , Baço/metabolismo , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA