Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Intervalo de ano de publicação
2.
Mol Med Rep ; 23(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33649841

RESUMO

Recent studies have reported that gene amplified in squamous cell carcinoma 1 (GASC1) is involved in the progression of several types of cancer. However, whether GASC1 promotes glioma progression remains unknown. Therefore, the present study aimed to investigate the effect of GASC1 exposure on glioma tumorigenesis. The western blot demonstrated that grade III and IV glioma tissues exhibited a higher mRNA and protein expression of GASC1. Moreover, CD133+ U87 or U251 cells from magnetic cell separation exhibited a higher GASC1 expression. Invasion Transwell assay, clonogenic assay and wound healing assay have shown that GASC1 inhibition using a pharmacological inhibitor and specific short hairpin (sh)RNA suppressed the invasive, migratory and tumorsphere forming abilities of primary culture human glioma cells. Furthermore, GASC1­knockdown decreased notch receptor (Notch) responsive protein hes family bHLH transcription factor 1 (Hes1) signaling. GASC1 inhibition reduced notch receptor 1 (NOTCH1) expression, and a NOTCH1 inhibitor enhanced the effects of GASC1 inhibition on the CD133+ U87 or U251 cell tumorsphere forming ability, while NOTCH1 overexpression abrogated these effects. In addition, the GASC1 inhibitor caffeic acid and/or the NOTCH1 inhibitor DAPT (a γ­Secretase Inhibitor), efficiently suppressed the human glioma xenograft tumors. Thus, the present results demonstrated the importance of GASC1 in the progression of glioma and identified that GASC1 promotes glioma progression, at least in part, by enhancing NOTCH signaling, suggesting that GASC1/NOTCH1 signaling may be a potential therapeutic target for glioma treatment.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Receptor Notch1/metabolismo , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Ácidos Cafeicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Diaminas/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/tratamento farmacológico , Glioma/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Camundongos Nus , Interferência de RNA , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/genética , Transdução de Sinais/genética , Tiazóis/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
Int J Mol Med ; 47(1): 113-124, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33155660

RESUMO

As hyperprolactinemia is observed in patients with bromocriptine­resistant prolactinoma, prolactin (PRL) has been implicated in the development of bromocriptine resistance. Since PRL primarily mediates cell survival and drug resistance via the Janus kinase­2 (JAK2)/signal transducer and activator of transcription 5A (STAT5) signaling pathway, the STAT5 inhibitor, pimozide, may inhibit cell proliferation and reverse bromocriptine resistance in prolactinoma cells. In the present study, compared with bromocriptine or pimozide alone, the combination of pimozide and bromocriptine exerted enhanced reduction in cell growth and proliferation, and increased apoptosis and cell cycle arrest in bromocriptine­resistant prolactinoma cells. A reduction in phospho­STAT5, cyclin D1 and B­cell lymphoma extra­large (Bcl­xL) expression levels were observed in cells treated with the combination of drugs. In addition, pimozide suppressed spheroid formation of human pituitary adenoma stem­like cells, and reduced the protein expression of the cancer stem cell markers, CD133 and nestin. Pimozide did not exert any additional antitumor activity in STAT5­knockdown primary culture cells of human bromocriptine­resistant prolactinomas. Furthermore, Pimozide combined with bromocriptine treatment significantly reduced human prolactinoma xenograft growth. Western blot and immunohistochemical analyses also demonstrated significant inhibition of cell proliferation and stem cell marker proteins in vivo. Collectively, these data indicated that pimozide treatment reduced prolactinoma growth by targeting both proliferating cells and stem cells, at least in part, by inhibiting the STAT5/Bcl­xL and STAT5/cyclin D1 signaling pathways.


Assuntos
Bromocriptina/farmacologia , Ciclina D1/metabolismo , Pimozida/farmacologia , Neoplasias Hipofisárias , Prolactinoma , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína bcl-X/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Prolactinoma/tratamento farmacológico , Prolactinoma/metabolismo , Prolactinoma/patologia , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Int J Med Sci ; 17(18): 3174-3189, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33173437

RESUMO

Prolactinomas are the most common type of functional pituitary adenoma. Although bromocriptine is the preferred first line treatment for prolactinoma, resistance frequently occurs, posing a prominent clinical challenge. Both the prolactin receptor (PRLR) and estrogen receptor α (ERα) serve critical roles in the development and progression of prolactinomas, and whether this interaction between PRLR and ERα contributes to bromocriptine resistance remains to be clarified. In the present study, increased levels of ERα and PRLR protein expression were detected in bromocriptine-resistant prolactinomas and MMQ cells. Prolactin (PRL) and estradiol (E2) were found to exert synergistic effects on prolactinoma cell proliferation. Furthermore, PRL induced the phosphorylation of ERα via the JAK2-PI3K/Akt-MEK/ERK pathway, while estrogen promoted PRLR upregulation via pERα. ERα inhibition abolished E2-induced PRLR upregulation and PRL-induced ERα phosphorylation, and fulvestrant, an ERα inhibitor, restored pituitary adenoma cell sensitivity to bromocriptine by activating JNK-MEK/ERK-p38 MAPK signaling and cyclin D1 downregulation. Collectively, these data suggest that the interaction between the estrogen/ERα and PRL/PRLR pathways may contribute to bromocriptine resistance, and therefore, that combination treatment with fulvestrant and bromocriptine (as opposed to either drug alone) may exert potent antitumor effects on bromocriptine-resistant prolactinomas.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Receptor alfa de Estrogênio/metabolismo , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Hipofisárias/terapia , Prolactinoma/terapia , Receptores da Prolactina/metabolismo , Adolescente , Adulto , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bromocriptina/farmacologia , Bromocriptina/uso terapêutico , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Estradiol/metabolismo , Receptor alfa de Estrogênio/análise , Receptor alfa de Estrogênio/antagonistas & inibidores , Feminino , Fulvestranto/farmacologia , Fulvestranto/uso terapêutico , Humanos , Hipofisectomia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Hipófise/patologia , Hipófise/cirurgia , Neoplasias Hipofisárias/patologia , Prolactina/metabolismo , Prolactinoma/patologia , Ratos , Receptores da Prolactina/análise , Adulto Jovem
5.
Huan Jing Ke Xue ; 40(1): 114-120, 2019 Jan 08.
Artigo em Chinês | MEDLINE | ID: mdl-30628265

RESUMO

Characterization of the size distribution and carbon components in particulates has become important for identifying the particulates in the atmosphere. The size distribution and carbon components of atmospheric particulate matter from motor vehicles in different regions were analyzed by using Micro-orifice uniform deposition impactors (MOUDI) and the organic carbon/elemental carbon (OC/EC) analyzer. With increasing particle size, the mass concentration of raw diesel/gasoline decreases. The highest mass concentration of particles collected near the chimney of an engine laboratory was observed for particle sizes ranging from 0.32-0.56 µm, while particles with sizes from 1.0-1.8 µm in the basement garage showed the most mass fractions. The OC1, OC2, and OC3 were the major parts of the OC contents in raw diesel particles. The EC2 was the main part of EC. The atmospheric particles collected in typical regions contained more OC3 and OC4. EC1 was the main part of EC in particles collected from the basement garage. The OC/EC ratios of raw diesel particles ranged from 0.92 to 2.50. The OC/EC ratios of particles collected near the chimney of an engine laboratory ranged from 1.40 to 2.53 and that of particles collected in the basement garage ranged from 2.36 to 4.82. Moreover, the OC/EC ratios in particles collected in the basement garage normally exceeded 2.0 and reached 4.82 at the largest size, which implies that many secondary particles were generated in the basement garage. The above-mentioned characteristics provide references that are beneficial for the identification of particulates in the atmosphere originating from motor vehicles.

6.
Biomed Pharmacother ; 98: 440-445, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29278854

RESUMO

Aristaless-like homeobox 4 (ALK4) is a member of ALK proteins family and plays an important role in tumorigenesis. However, the expression and function of ALK4 in glioma remain largely unknown. The aim of our study was to elucidate its expression pattern in human glioma tissues and cell lines, as well as its functions in glioma cells. Our results demonstrated that ALK4 was lowly expressed in human glioma tissues and cell lines. Additionally, overexpression of ALK4 significantly suppressed the proliferation, migration and invasion of glioma cells, as well as inhibited the epithelial-mesenchymal transition (EMT) phenotype in glioma cells. Furthermore, overexpression of ALK4 significantly downregulated the phosphorylation levels of JAK2 and STAT3 in U87 cells. STAT3 inhibitor (Niclosamide) obviously enhanced ALK4-inhibted glioma cell proliferation and invasion. In conclusion, we demonstrated that overexpression of ALK4 suppressed glioma cell proliferation, migration and invasion through the inactivation of JAK/STAT3 signaling pathway. Thus, ALK4 may be a potential therapeutic target for the treatment of glioma.


Assuntos
Receptores de Ativinas Tipo I/genética , Movimento Celular/genética , Proliferação de Células/genética , Glioma/genética , Janus Quinase 2/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Invasividade Neoplásica/genética
7.
J Exp Clin Cancer Res ; 36(1): 6, 2017 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-28061785

RESUMO

BACKGROUND: Although chemopreventative agents targeting the estrogen/estrogen receptor (ER) pathway have been effective for ER+ breast cancers, prevention of hormone receptor-negative breast cancers, such as Her2/erbB-2+ breast cancers, remains a significant issue. Previous studies have demonstrated that administration of EGFR/erbB-2-targeting lapatinib to MMTV-erbB-2 transgenic mice inhibited mammary tumor development. The prevention, however, was achieved by prolonged high dose exposure. The tolerance to high dose/long-term drug administration may hinder its potential in clinical settings. Therefore, we aimed to test a novel, short-term chemopreventative strategy using lapatinib during the premalignant risk window in MMTV-erbB-2 mice. METHODS: We initially treated cultured cells with lapatinib to explore the anti-proliferative effects of lapatinib in vitro. We used a syngeneic tumor graft model to begin exploring the in vivo anti-tumorigenic effects of lapatinib in MMTV-erbB-2 mice. Then, we tested the efficacy of brief exposure to lapatinib (100 mg/kg/day for 8 weeks), beginning at 16 weeks of age, in the prevention of mammary tumor development in MMTV-erbB-2 mice. RESULTS: In the syngeneic tumor transplant model, we determined that lapatinib significantly inhibited tumor cell proliferation. Furthermore, we demonstrated that short-term lapatinib exposure resulted in life-long protective effects, as supported by increased tumor latency in lapatinib-treated mice compared to the control mice. We further established that delayed tumor development in the treated mice was preceded by decreased BrdU nuclear incorporation and inhibited mammary morphogenesis. Molecular analysis indicated that lapatinib inhibited phosphorylation and expression of EGFR, erbB-3, erbB-2, Akt1, and Erk1/2 in premalignant mammary tissues. Also, lapatinib drastically inhibited the phosphorylation and expression of ERα and the transcription of ER target genes in premalignant mammary tissues. We also determined that lapatinib suppressed the stemness of breast cancer cell lines, as evidenced by decreased tumorsphere formation and ALDH+ cell populations. CONCLUSIONS: Taken together, these data demonstrate that brief treatment with EGFR/erbB-2-targeting agents before the onset of tumors may provide lifelong protection from mammary tumors, through the concurrent inhibition of erbB-2 and ER signaling pathways and consequential reprogramming. Our findings support further clinical testing to explore the benefit of shorter lapatinib exposure in the prevention of erbB-2-mediated carcinogenesis.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Mamárias Experimentais/prevenção & controle , Quinazolinas/administração & dosagem , Receptor ErbB-2/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Esquema de Medicação , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lapatinib , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/farmacologia , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo
8.
Phytother Res ; 30(7): 1095-103, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27145435

RESUMO

This meta-analysis aimed to evaluate the immunomodulating function of Yupingfeng Formula (YPFF) in children with recurrent respiratory tract infections (RRTIs). The PubMed, EMBASE, Cochrane Library, CNKI and WanFang databases were searched for randomized controlled trials comparing with and without YPFF for RRTIs in children. Twelve trials with 1236 patients were identified. Adjuvant treatment with YPFF significantly increased serum levels of IgA (weighted mean difference [WMD] 0.33 mg/mL; 95% confidence interval [CI] 0.20 to 0.45), IgG (WMD 1.36 mg/mL; 95% CI 1.06 to 1.65), IgM (WMD 0.16 mg/mL; 95% CI 0.02 to 0.31), and CD3(+) T-lymphocytes (WMD 10.16%; 95% CI 4.62 to 15.69) but not CD4(+) T-lymphocytes (WMD 3.16%; 95% CI -0.27 to 6.59) and CD8(+) T-lymphocytes (WMD -0.84%; 95% CI -2.50 to 0.81). YPFF also reduced the frequency of RRTIs (WMD -3.80 times; 95% CI -4.86 to -2.74) and increased total effective rates of symptom improvement (risk ratio: 1.44; 95% CI 1.19 to 1.75). Adjuvant treatment with YPFF could improve total clinical effective rate and decrease the frequency of respiratory tract infections in children with RRTIs. The beneficial effects of YPFF may be correlated to its immunomodulating action. More well-designed trials with larger sample sizes are needed to evaluate its efficacy and safety. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Infecções Respiratórias/tratamento farmacológico , Criança , Medicamentos de Ervas Chinesas/efeitos adversos , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Recidiva
9.
Int J Mol Med ; 30(2): 277-82, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22664747

RESUMO

Stress-dose of glucocorticoid has been demonstrated to be beneficial for trauma patients in clinical studies. Recently, a heterogeneous population of myeloid cells with immunosuppressive activity named myeloid-derived suppressor cells (MDSCs) has been found to accumulate in the trauma host and can be induced by glucocorticoids in vitro. In order to explore the effect of endogenous glucocorticoids on MDSCs under trauma conditions, we blocked the glucocorticoid signal in a murine trauma model using the antagonist of the glucocorticoid receptor RU486 (mifepristone). We found for the first time that RU486 not only blunted MDSC expansion induced by trauma in the spleen, peripheral blood and bone marrow especially at 6 h after traumatic stress but also decreased the survival rate from 100 to 20% in traumatic mice within 7 days. Moreover, neither MDSCs producing arginase-1 nor the morphological characterization of trauma-induced MDSCs was affected by the blockage of the glucocorticoid receptor. Our results suggest that endogenous glucocorticoids may promote MDSCs expansion in a murine trauma model and MDSCs may be beneficial for the trauma host.


Assuntos
Glucocorticoides/metabolismo , Células Mieloides/metabolismo , Ferimentos e Lesões/metabolismo , Animais , Arginase/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Antígeno CD11b/metabolismo , Células Cultivadas , Glucocorticoides/antagonistas & inibidores , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mifepristona/farmacologia , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Baço/efeitos dos fármacos , Baço/metabolismo , Ferimentos e Lesões/mortalidade
10.
Tohoku J Exp Med ; 226(4): 267-74, 2012 04.
Artigo em Inglês | MEDLINE | ID: mdl-22481303

RESUMO

Glioblastoma multiforme is an aggressive brain tumor with a poor prognosis. The glioblastoma stem-like cells (GSCs) represent a rare fraction of human glioblastoma cells with the capacity for multi-lineage differentiation, self-renewal and exact recapitulation of the original tumor. Interestingly, GSCs are more radioresistant compared with other tumor cells. In addition, the remarkable radioresistance of GSCs has been known to promote radiotherapy failure and therefore is associated with a significantly higher risk of a local tumor recurrence. Moreover, the hyperactive cell cycle checkpoint kinase (Chk) 1 and 2 play a pivotal role in the DNA damage response including radiation and chemical therapy. Based on aforementioned, we hypothesized that knockdown of Chk1 or Chk2 might confer radiosensitivity on GSCs and thereby increases the efficiency of radiotherapy. In this study, we knocked down the expression of Chk1 or Chk2 in human GSCs using lentivirus-delivered short hairpin RNA (shRNA) to examine its effect on the radiosensitivity. After radiation, the apoptosis rate and the cell cycle of GSCs were measured with Flow Cytometry. Compared with control GSCs (apoptosis, 7.82 ± 0.38%; G2/M arrest, 60.20 ± 1.28%), Chk1 knockdown in GSCs increased the apoptosis rate (37.87 ± 0.32%) and decreased the degree of the G2/M arrest (22.37 ± 2.01%). In contrast, the radiosensitivity was not enhanced by Chk2 knockdown in GSCs. These results suggest that depletion of Chk1 may improve the radio-sensitivity of GSCs via inducing cell apoptosis. In summary, the therapy targeting Chk1 gene in the GSCs may be a novel way to treat glioblastoma.


Assuntos
Neoplasias Encefálicas/radioterapia , Glioblastoma/radioterapia , Células-Tronco Neoplásicas , Proteínas Quinases/genética , Tolerância a Radiação/genética , Apoptose/fisiologia , Apoptose/efeitos da radiação , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Terapia Combinada , Técnicas de Silenciamento de Genes , Terapia Genética/métodos , Glioblastoma/enzimologia , Glioblastoma/patologia , Humanos , Lentivirus/genética , Células-Tronco Neoplásicas/enzimologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia
11.
J Huazhong Univ Sci Technolog Med Sci ; 32(1): 75-81, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22282249

RESUMO

The osteogenic in vitro effect of low intensity pulsed ultrasound (LIPUS) on SD rat adipose-derived stem cells (ADSCs) was investigated. Rat ADSCs underwent LIPUS (intensity=100 mW/cm(2)) or sham exposure for 8 min per treatment once everyday in vitro, and then the alkaline phosphatase (ALP) activity and mineralized nodule formation were assessed to evaluate the osteogenic effect of LIPUS on ADSCs. To further explore the underlying mechanism, the osteogenic-related gene mRNA expression was determined by using reverse transcriptase-polymerase chain reaction (RT-PCR) at 1st, 3rd, 5th, 7th day after exposure repectively. Westen blot was used to evaluate the protein expression levels of two osteogenic differentiation associated genes at 7th and 14th day repectively. It was found that ALP activity was increased after LIPUS exposure and LIPUS resulted in mineralized nodule formation of ADSCs in vitro. LIPUS-treated ADSCs displayed higher mRNA expression levels of runt-related transcription factor 2 (Runx2), osteocalcin (OCN), ALP and bone sialoprotein (BSP) genes than controls, and the protein levels of Runx2 and BSP were also increased. The results suggested that LIPUS may induce the osteogenic differentiation of ADSCs in vitro.


Assuntos
Adipócitos/citologia , Osteoblastos/citologia , Osteogênese/fisiologia , Sonicação/métodos , Células-Tronco/citologia , Adipócitos/fisiologia , Adipócitos/efeitos da radiação , Animais , Células Cultivadas , Ondas de Choque de Alta Energia , Osteoblastos/fisiologia , Osteoblastos/efeitos da radiação , Osteogênese/efeitos da radiação , Doses de Radiação , Ratos , Ratos Sprague-Dawley , Células-Tronco/fisiologia , Células-Tronco/efeitos da radiação
12.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-248558

RESUMO

The osteogenic in vitro effect of low intensity pulsed ultrasound (LIPUS) on SD rat adipose-derived stem cells (ADSCs) was investigated.Rat ADSCs underwent LIPUS (intensity=100 mW/cm2) or sham exposure for 8 min per treatment once everyday in vitro,and then the alkaline phosphatase (ALP) activity and mineralized nodule formation were assessed to evaluate the osteogenic effect of LIPUS on ADSCs.To further explore the underlying mechanism,the osteogenic-related gene mRNA expression was determined by using reverse transcriptase-polymerase chain reaction (RT-PCR) at 1st,3rd,5th,7th day after exposure repectively.Westen blot was used to evaluate the protein expression levels of two osteogenic differentiation associated genes at 7th and 14th day repectively.It was found that ALP activity was increased after LIPUS exposure and LIPUS resulted in mineralized nodule formation of ADSCs in vitro.LIPUS-treated ADSCs displayed higher mRNA expression levels of runt-related transcription factor 2 (Runx2),osteocalcin (OCN),ALP and bone sialoprotein (BSP) genes than controls,and the protein levels of Runx2 and BSP were also increased.The results suggested that LIPUS may induce the osteogenic differentiation of ADSCs in vitro.

13.
Oncol Rep ; 26(6): 1457-64, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21822544

RESUMO

The hypoxia inducible factor 1 α (HIF-1α) activity has been associated with various hemorrhagic events. The biological role of HIF-1α in the hemorrhagic transformation of pituitary adenomas remains unknown. We hypothesized that fast growing tumor cells tend to predispose themselves to sublethal hypoxia and activate the HIF-1α signaling pathway, leading to hemorrhagic transformation in pituitary adenomas. Here, we used apoplectic and non-apoplectic pituitary adenomas to determine the involvement of HIF-1α signaling in intratumoral hemorrhage. We employed HIF-1α overexpression/knockdown strategies to examine the association between HIF-1α signaling and hemorrhagic presentation in vitro and in vivo. In support of our hypothesis, compared with non-hemorrhagic pituitary adenomas, higher cellular proliferation was observed in hemorrhagic ones and it correlated with increased HIF-1α signaling. HIF-1α overexpression activated its downstream genes, vascular endothelial growth factor and the proapoptotic BNIP3, in MMQ pituitary adenoma cells and this up-regulation was attenuated by HIF-1 siRNA. In vivo studies using MMQ cell xenografts in nude mice showed that HIF-1α overexpression significantly promoted hemorrhagic transformation. Our study indicates that tumor hypoxia, following rapid tumor growth, may promote hemorrhagic transformation in pituitary adenomas via the HIF-1α signaling pathway.


Assuntos
Adenoma/patologia , Hipóxia Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hemorragias Intracranianas/patologia , Neoplasias Hipofisárias/patologia , Transdução de Sinais , Adenoma/metabolismo , Adolescente , Adulto , Animais , Apoptose , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Transplante de Neoplasias , Neoplasias Hipofisárias/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Ativação Transcricional , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
14.
Int J Mol Sci ; 12(6): 4165-79, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21747731

RESUMO

Pituitary apoplexy is a clinical syndrome with unknown pathogenesis. Therefore, identifying the underlying mechanisms is of high clinical relevance. Tumor necrosis factor alpha (TNF-α) is a critical cytokine mediating various hemorrhagic events, but little is known about its involvement in pituitary apoplexy. Here we show that TNF-α may be an important regulator of hemorrhagic transformation in pituitary adenomas. In this study, sixty surgical specimens of hemorrhagic and non-hemorrhagic human pituitary adenomas were examined. Hemorrhagic pituitary adenomas displayed higher protein and mRNA levels of TNF-α, vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) compared with those of non-hemorrhagic tumors. Exposure of MMQ pituitary adenoma cells to TNF-α induced VEGF and MMP-9 expression in vitro. Additionally, TNF-α administration caused hemorrhagic transformation and enhanced VEGF and MMP-9 expression in MMQ pituitary adenoma cell xenografts in mice. Blockers of VEGF or MMP-9, either alone or in combination, attenuated but not abrogated TNF-α mediated hemorrhagic transformation in xenografts. This study suggests that TNF-α may play a role in the development of intratumoral hemorrhage in pituitary adenomas via up-regulation of VEGF and MMP-9.


Assuntos
Adenoma/patologia , Regulação Neoplásica da Expressão Gênica , Hemorragia , Metaloproteinase 9 da Matriz/metabolismo , Neoplasias Hipofisárias/patologia , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenoma/metabolismo , Adolescente , Adulto , Inibidores da Angiogênese/farmacologia , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Bevacizumab , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hemorragia/genética , Humanos , Masculino , Metaloproteinase 9 da Matriz/química , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Hipofisárias/metabolismo , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Transplante Heterólogo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...