Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Theranostics ; 9(3): 796-810, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30809309

RESUMO

Background and Aims: Cancer cells prefer aerobic glycolysis to maintain growth advantages, but the role of long non-coding RNAs (lncRNAs) in glycometabolism still remains unclear. Here we identified one cytoplasmic lncRNA LINC01554 as a significantly downregulated lncRNA in hepatocellular carcinoma (HCC) and aimed to investigate its role in cellular glucose metabolism in the development and progression of HCC. Methods: Quantitative real-time PCR was used to determine the expression level of LINC01554. Downregulation of LINC01554 by miR-365a at transcriptional level was assessed by luciferase reporter assay. Subcellular fractionation assay and RNA fluorescence in situ hybridization were performed to detect the subcellular localization of LINC01554. RNA pull-down assay, mass spectrometry, and RNA immunoprecipitation assay were used to identify the underlying molecular mechanisms. The tumor-suppressive function of LINC01554 was determined by both in vitro assay and nude mice xenograft model. Results: LINC01554 was frequently downregulated in HCC, which was significantly associated with tumor invasion (P = 0.005), tumor size (P = 0.041), tumor staging (P = 0.023) and shorter survival (P = 0.035) of HCC patients. Luciferase reporter assay unraveled that LINC01554 was negatively regulated by miR-365a. Subcellular fractionation assay and RNA FISH revealed the cytoplasmic predominance of LINC01554 in MIHA cells and HCC clinical samples. Ectopic expression of LINC01554 inhibited HCC cell growth, colony formation in soft agar, foci formation, and tumor formation in nude mice. LINC01554 promoted the ubiquitin-mediated degradation of PKM2 and inhibited Akt/mTOR signaling pathway to abolish aerobic glycolysis in HCC cells. Further study found that LINC01554-knockout could effectively reverse the tumor-suppressive effect of LINC01554. Conclusions: Our results identify LINC01554 as a novel tumor suppressor in HCC and unravel its underlying molecular mechanism in reprogramming cellular glucose metabolism. LINC01554 could possibly serve as a novel prognostic biomarker and provide the rationale for HCC therapy.


Assuntos
Carcinoma Hepatocelular/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glucose/metabolismo , Neoplasias Hepáticas/metabolismo , Piruvato Quinase/genética , RNA Longo não Codificante/genética , Animais , Carcinoma Hepatocelular/genética , Regulação para Baixo , Feminino , Humanos , Neoplasias Hepáticas/genética , Masculino , Camundongos Endogâmicos BALB C , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Serina-Treonina Quinases TOR/antagonistas & inibidores
3.
Proc Natl Acad Sci U S A ; 115(38): E8948-E8957, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30158168

RESUMO

Angiogenesis is essential in the early stage of solid tumor recurrence, but how a suspensive tumor is reactivated before angiogenesis is mostly unknown. Herein, we stumble across an interesting phenomenon that s.c. xenografting human lung cancer tissues can awaken the s.c. suspensive tumor in nude mice. We further found that a high level of insulin-like growth factor 1 (IGF1) was mainly responsible for triggering the transition from suspensive tumor to progressive tumor in this model. The s.c. suspensive tumor is characterized with growth arrest, avascularity, and a steady-state level of proliferating and apoptotic cells. Intriguingly, CD133+ lung cancer stem cells (LCSCs) are highly enriched in suspensive tumor compared with progressive tumor. Mechanistically, high IGF1 initiates LCSCs self-renewal from asymmetry to symmetry via the activation of a PI3K/Akt/ß-catenin axis. Next, the expansion of LCSC pool promotes angiogenesis by increasing the production of CXCL1 and PlGF in CD133+ LCSCs, which results in lung cancer recurrence. Clinically, a high level of serum IGF1 in lung cancer patients after orthotopic lung cancer resection as an unfavorable factor is strongly correlated with the high rate of recurrence and indicates an adverse progression-free survival. Vice versa, blocking IGF1 or CXCL1/PlGF with neutralizing antibodies can prevent the reactivation of a suspensive tumor induced by IGF1 stimulation in the mouse model. Collectively, the expansion of LCSC pool before angiogenesis induced by IGF1 is a key checkpoint during the initiation of cancer relapse, and targeting serum IGF1 may be a promising treatment for preventing recurrence in lung cancer patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias Pulmonares/patologia , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/patologia , Antígeno AC133/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/sangue , Linhagem Celular Tumoral , Proliferação de Células , Quimiocina CXCL1/antagonistas & inibidores , Quimiocina CXCL1/metabolismo , Feminino , Humanos , Fator de Crescimento Insulin-Like I/análise , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Neoplasias Pulmonares/sangue , Camundongos , Camundongos Nus , Recidiva Local de Neoplasia/sangue , Neovascularização Patológica/sangue , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Crescimento Placentário/antagonistas & inibidores , Fator de Crescimento Placentário/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo
4.
Int J Biol Sci ; 13(4): 471-479, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28529455

RESUMO

Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis, accounting for approximately 12-24% of breast cancer cases. Accumulating evidence has indicated that there is no effective targeted therapy available for TNBC. Dipalmitoylphosphatidic acid (DPPA) is a bioactive phospholipid. However, the function of DPPA in the growth of TNBC has not yet been studied. In this study, we employed TNBC cells and a subcutaneous tumor model to elucidate the possible effect of DPPA on tumor growth in TNBC. We showed that DPPA significantly inhibited tumor growth in the mouse subcutaneous tumor model and suppressed cell proliferation and angiogenesis in TNBC tumor tissues. This inhibition was mediated partly by suppressing the expression of cyclin B1 (CCNB1), which directly promoted the accumulation of cells in the G2 phase and arrested cell cycle progression in human TNBC. In addition, the inhibition of tumor growth by DPPA may also be mediated by the suppression of tumor angiogenesis in TNBC. This work provides initial evidence that DPPA might be vital as an anti-tumor drug to treat TNBC.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ácidos Fosfatídicos/farmacologia , Ácidos Fosfatídicos/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Fase G2/efeitos dos fármacos , Fase G2/genética , Humanos , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Neurosci Lett ; 631: 85-90, 2016 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-27521753

RESUMO

Traumatic brain injury (TBI) is the leading cause of mortality and disability among male adolescents and young adults; and mild traumatic brain injury is the most common type of traumatic brain injury. The disruption of blood-brain barrier (BBB) plays an important role in brain trauma. Previously, we have found that slit2, a member of slit protein family, increases permeability of BBB. In the present study, we examined the role of slit2 in the pathogenesis of mild TBI in a mouse model of micro TBI. Rhodamine BandPI (PropidiumIodide) staining were used to detect the permeability of BBB and cell death, respectively. The leakage of Rhodamine B and cell death were significantly increased in Slit2-Tg mice than in C57 control mice after micro TBI. The present results suggest that over expression of slit2 plays a detrimental role in the pathophysiology of mild TBI.


Assuntos
Barreira Hematoencefálica/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Morte Celular , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
6.
Ann Anat ; 200: 79-87, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25819501

RESUMO

Many human disorders induce high salinity in tissues and organs, interfering with their normal physiological functions. Using a mouse model, we demonstrated that high salt intake caused infertility. Specifically, we established that high salinity dramatically affects ovarian follicle development and the extent of follicular atresia. However, it did not significantly influence the primordial follicles. TUNEL assays revealed that high salt intake inhibited follicle development by inducing the granulosa and theca cells that surround the oocytes to undergo apoptosis. Furthermore, immunohistological staining for the proliferation markers Ki67 and PH3 showed that high salt intake also repressed granulosa cell proliferation. In vitro testing of granulosa cells also confirmed that high salt significantly repressed cell proliferation and promoted cell apoptosis. In summary, high salt consumption negatively impacts reproductive functions in female mice by interfering with ovarian folliculogenesis.


Assuntos
Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , Cloreto de Sódio/toxicidade , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Atresia Folicular/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Infertilidade Feminina/induzido quimicamente , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/efeitos dos fármacos , Gravidez , Cultura Primária de Células , Células Tecais/efeitos dos fármacos
7.
Cell Cycle ; 14(5): 772-83, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25738919

RESUMO

Both pre-gestational and gestational diabetes have an adverse impact on heart development, but little is known about the influence on the early stage of heart tube formation. Using early gastrulating chick embryos, we investigated the influence of high glucose on the process of heart tube formation, specifically during the primary heart field phase. We demonstrated that high-glucose exposure resulted in 3 types of heart tube malformation: 1) ventricular hypertrophy, 2) ventricular hypertrophy with dextrocardia and 3) ventricular hypertrophy and dextrocardia with the fusion anomaly of a bilateral primary heart tube. Next, we found that these malformation phenotypes of heart tubes might mainly originate from the migratory anomaly of gastrulating precardiac mesoderm cells rather than cell proliferation in the developmental process of bilateral primary heart field primordia. The treatment of rapamycin (RAPA), an autophagy inducer, led to a similar heart tube malformation phenotype as high glucose. Additionally, high-glucose exposure promoted the expression of the key autophagy protein LC3B in early chick tissue. Atg7 is strongly expressed in the fusion site of bilateral primary heart tubes. All of these data imply that autophagy could be involved in the process of high-glucose-induced malformation of the heart tube.


Assuntos
Autofagia/efeitos dos fármacos , Glucose/farmacologia , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/patologia , Coração/embriologia , Animais , Movimento Celular/efeitos dos fármacos , Embrião de Galinha , Gástrula/efeitos dos fármacos , Gástrula/patologia , Gastrulação/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Coração/efeitos dos fármacos , Mesoderma/efeitos dos fármacos , Mesoderma/patologia , Organogênese/efeitos dos fármacos , Fenótipo , Sirolimo/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
8.
Oncotarget ; 6(5): 3123-35, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25605242

RESUMO

Slit2 is often overexpressed in cancers. Slit2 is a secreted protein that binds to Roundabout (Robo) receptors to regulate cell growth and migration. Here, we employed several complementary mouse models of intestinal cancers, including the Slit2 transgenic mice, the ApcMin/+ spontaneous intestinal adenoma mouse model, and the DMH/DSS-induced colorectal carcinoma model to clarify function of Slit2/Robo1 signaling in intestinal tumorigenesis. We showed that Slit2 and Robo1 are overexpressed in intestinal tumors and may contribute to tumor generation. The Slit2/Robo1 signaling can induce precancerous lesions of the intestine and tumor progression. Ectopic expression of Slit2 activated Slit2/Robo1 signaling and promoted tumorigenesis and tumor growth. This was mediated in part through activation of the Src signaling, which then down-regulated E-cadherin, thereby activating Wnt/ß-catenin signaling. Thus, Slit2/Robo1 signaling is oncogenic in intestinal tumorigenesis.


Assuntos
Adenoma/enzimologia , Carcinoma/enzimologia , Neoplasias Colorretais/enzimologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Intestinal/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Quinases da Família src/metabolismo , Adenoma/genética , Adenoma/patologia , Animais , Carcinoma/genética , Carcinoma/patologia , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Genes APC , Células HCT116 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mucosa Intestinal/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Interferência de RNA , Receptores Imunológicos/genética , Transfecção , Carga Tumoral , Proteínas Roundabout
9.
J Alzheimers Dis ; 43(2): 535-48, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25114073

RESUMO

Alzheimer's disease (AD) is a progressive neurological disorder that primarily affects memory, and its prevalence is rising. Increasing evidence suggests that dysfunction of the blood-brain barrier (BBB) may be involved in AD and other neurodegenerative diseases. Herein, we report that the permeability of the BBB is increased and that AD-like alterations are present in Slit-2 overexpressing transgenic mice. We found that behavioral change and the corresponding molecular diagnostic markers of AD, such as hippocampal neuron apoptosis, amyloid-ß (Aß) protein deposition, and acetylcholinesterase expression, were increased in the Slit-2 transgenic mice. Moreover, the endothelial cells were dysfunctional, the size of the lateral ventricle cavity increased, and the permeability of the BBB increased. Additionally, there was an increased serum level of glutamate indicating that the BBB is related to AD. Finally, histopathological analysis of other organs in the Slit-2 overexpressing mice did not show any marked abnormalities. These findings demonstrate that Slit2 overexpression may be responsible for AD-like alterations and the increased BBB permeability in these mice. Our study provides a potential novel mechanism for the development of AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Barreira Hematoaquosa/fisiopatologia , Permeabilidade Capilar/genética , Regulação da Expressão Gênica/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas do Tecido Nervoso/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/sangue , Precursor de Proteína beta-Amiloide/genética , Animais , Barreira Hematoaquosa/ultraestrutura , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Ressonância Magnética Nuclear Biomolecular
10.
Melanoma Res ; 24(6): 545-55, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25244079

RESUMO

The TLR4/NF-κB signaling pathway plays a critical role in tumor progression. Andrographolide (Andro) has been reported to have anticancer activity in multiple types of cancer. However, the pharmacological activities of Andro in melanoma are not completely understood. In this study, we defined the anticancer effects of Andro in melanoma and elucidated its potential mechanisms of action. Our experiments showed that Andro significantly inhibited melanoma tumor growth and metastasis by inducing cell cycle arrest and apoptosis. In addition, Andro significantly inhibited the TLR4/NF-κB signaling pathway. Furthermore, the inactivation of TLR4/NF-κB signaling inhibited the mRNA and protein expression of CXCR4 and Bcl-6, which are antitumor genes. This work provides evidence that the TLR4/NF-κB signaling pathway is a potential therapeutic target and may also be indispensable in the Andro-mediated anticancer effect in melanoma.


Assuntos
Anti-Inflamatórios/farmacologia , Proliferação de Células/efeitos dos fármacos , Diterpenos/farmacologia , Melanoma Experimental/patologia , Neoplasias Cutâneas/patologia , Animais , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Feminino , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/metabolismo , Carga Tumoral/efeitos dos fármacos
11.
Mol Med Rep ; 10(3): 1513-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24938315

RESUMO

Podoplanin (PDPN) is a well established lymphatic endothelial marker and has frequently been observed in cancer cells at the edge of cancer masses. Previous studies investigating the association between PDPN expression and patient prognosis have had contradictory results. In the present study, it was hypothesized that the different locations of PDPN­positive cells may explain these varying results. The present study aimed to focus on PDPN expression at the edge of esophageal cancer cell nests. In order to analyze the clinical significance of this PDPN expression, immunohistochemistry was performed using esophageal cancer tissue microarrays. PDPN expression at the edge of the cancer cell nest was found to be significantly associated with invasion (P<0.05) and poor prognosis (P<0.001) in patients with cancer. To further investigate the role of PDPN expression in cancer cells, the PDPN gene was cloned and transfected into esophageal squamous cell carcinoma (ESCC) cell lines. PDPN expression was also knocked down using small interfering RNA. PDPN­positive cancer cells were found to exhibit invasion characteristics. Thus, PDPN expression at the edge of a cancer cell nest may indicate invasion and represent a poor prognostic factor for ESCCs.


Assuntos
Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/genética , Regulação Neoplásica da Expressão Gênica , Glicoproteínas de Membrana/metabolismo , Invasividade Neoplásica/diagnóstico , Invasividade Neoplásica/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Carcinoma de Células Escamosas do Esôfago , Humanos , Imuno-Histoquímica , Glicoproteínas de Membrana/genética , Pessoa de Meia-Idade , Prognóstico , RNA Interferente Pequeno , Transfecção
12.
Int J Biol Sci ; 10(4): 404-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24719558

RESUMO

Insulinomas are rare tumors, and approximately 10% of insulinomas are malignant. Accumulating evidence has implicated that we still lack effective therapy to treat the patients who are diagnosed with rare malignant insulinoma. Previous studies have reported that Andrographolide (Andro) could inhibit cell cycle progression, reduce cell invasion and induce cell apoptosis in many common cancer cells. However, the effects of andro are cell type-dependent. So we emplored the ß-TC-6 cells and the RIP1-Tag2 transgenic mouse model of endogenously growing insulinoma model to elucidate the possible anti-cancer effect of Andro on insulinoma, an uncommon type of malignant cancers in this study. Our experiments revealed that Andro significantly inhibited tumor growth at both the early-stage and the advanced-stage of insulinoma through targeting the TLR4/NF-κB signaling pathway. This work initially provides the evidence that the TLR4/NF-κB signaling pathway might be vital as a potential therapeutic target, and also indispensable in Andro-mediated anti-cancer effect in insulinoma.


Assuntos
Antineoplásicos/uso terapêutico , Diterpenos/uso terapêutico , Insulinoma/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Diterpenos/farmacologia , Humanos , Insulinoma/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neovascularização Patológica/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo
13.
PLoS One ; 9(3): e91320, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24632801

RESUMO

Studies have indicated that the aggregation of activated platelets with cancer cells facilitates tumor metastasis; the adhesion molecule P-selectin may be an important mediator of this process, but the detailed mechanism is unclear. In the current study, we established a B16F10 (B16) cell metastatic model in P-selectin knockout (P-sel-/-) mice to determine the effect of P-selectin-mediated platelet adhesion on metastasis. Compared with C57 mice, P-sel-/- mice developed fewer metastatic foci, and cell proliferation within the metastatic tumors was inhibited by P-selectin deficiency. The platelet refusion assay demonstrated that mice with P-sel-/- platelets developed fewer lung metastatic foci (P<0.01) with a lower microvascular density (MVD) than mice with wild-type platelets. A co-culture model of platelets and B16 cells was utilized to determine the difference in VEGF concentration in the supernatants. The results demonstrated that the supernatant from the P-sel-/- platelet/B16 co-culture had a lower concentration of VEGF. Therefore, our findings indicated that P-selectin deficiency inhibited the metastasis of B16 cells and that wild-type platelet refusion reversed this inhibition. The P-selectin-mediated interaction between platelets and B16 cells promoted angiogenesis by up-regulating VEGF.


Assuntos
Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Selectina-P/metabolismo , Adesividade Plaquetária , Animais , Proliferação de Células , Modelos Animais de Doenças , Melanoma Experimental/genética , Camundongos , Camundongos Knockout , Metástase Neoplásica , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Selectina-P/genética , Adesividade Plaquetária/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Int J Biol Sci ; 10(2): 136-41, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24520211

RESUMO

Rip1-Tag2 mice is one overt pancreatic ß-cell tumor model, which is widely used for studying pancreas tumor angiogenesis and tumor development. However, tumor metastasis in Rip1-Tag2 mice had rarely been reported, in this present study, we find some micrometastasis in lung and spleen of the Rip1-Tag2 mice at advanced stage, which is important for uncovering metastasis cell characteristics and exploring how to survive in cancer microenvironment. To study the micrometastasis of Rip1-Tag2 mice in advanced pancreatic cancer, we first observed the pathology process of ß cell tumor in Rip1-Tag2 mice through HE staining, then we performed immunohistochemistry with insulin antibody, T-antigen antibodies and C-petide antibody on lung and spleen tissues sections from advanced stage, comparing with background wild-type C57BL/6 mice sections. The results indicated that micrometastasis expressing insulin was found in the Rip1-Tag2 mice lung, and spleen. Further evidences demonstrate pathology structure of lung and spleen are damaged. Interestingly and importantly, the expression of T antigen and insulin antibodies are all decreased in advanced stage of primary ß cell tumor, which suggest that the at least partly micrometastasis is derived from the early stage or from advanced stage of ß cell tumor then return to undifferentiated state like cancer stem cell. The findings contributed to the study of cancer metastasis and cancer stem cell.


Assuntos
Proteínas Ativadoras de GTPase/genética , Insulina/metabolismo , Insulinoma/genética , Pulmão/metabolismo , Baço/metabolismo , Animais , Antígenos Virais de Tumores/metabolismo , Regulação para Baixo , Insulinoma/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Micrometástase de Neoplasia , Baço/patologia , Microambiente Tumoral
15.
Mol Med Rep ; 8(6): 1734-40, 2013 12.
Artigo em Inglês | MEDLINE | ID: mdl-24068303

RESUMO

Angiogenesis exhibits a significant effect on tumor progression. Inhibiting angiogenesis may provide significant advantages over currently available therapeutics for cancer therapies thus, the development of a system of screening angiogenesis is essential. In the present study, a novel available system of screening angiogenesis inhibitors by four steps was developed. The chorioallantoic membrane (CAM), yolk sac membrane and early chick embryo blood island assay were initially performed to obtain possible antitumor compounds. The MMTV­PyMT transgenic breast cancer mouse model was used for final screening and to confirm potential antitumor effects. Four angiogenesis inhibitors were isolated from 480 compounds, which were obtained from ICCB known bioactives library, by a combination of the CAM, yolk sac membrane and early chick embryo blood island assay. The MMTV­PyMT mouse was treated with one of four agents and it was demonstrated that the tumor volume was significantly inhibited. These results demonstrate that the four­step screening system is feasible.


Assuntos
Inibidores da Angiogênese/análise , Inibidores da Angiogênese/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores da Angiogênese/uso terapêutico , Animais , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/efeitos dos fármacos , Membrana Corioalantoide/patologia , Dimetil Sulfóxido/farmacologia , Modelos Animais de Doenças , Feminino , Ensaios de Triagem em Larga Escala , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Membranas/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Saco Vitelino/irrigação sanguínea , Saco Vitelino/efeitos dos fármacos
16.
PLoS One ; 8(4): e60027, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23560067

RESUMO

BACKGROUND: Deletion of 3p is one of the most frequent genetic alterations in esophageal squamous cell carcinoma (ESCC), suggesting the existence of one or more tumor suppressor genes (TSGs) within these regions. In this study, one TSG, CACNA2D3 at 3p21.1, was characterized. METHODS: Expression of CACNA2D3 in ESCCs was tested by quantitative real-time PCR and tissue microarray. The mechanism of CACNA2D3 downregulation was investigated by methylation-specific polymerase chain reaction (MS-PCR). The tumor suppressive function of CACNA2D3 was characterized by both in vitro and in vivo tumorigenic assays, cell migration and invasion assays. RESULTS: CACNA2D3 was frequently downregulated in ESCCs (24/48, 50%), which was significantly associated with promoter methylation and allele loss (P<0.05). Tissue microarray result showed that downregulation of CACNA2D3 was detected in (127/224, 56.7%) ESCCs, which was significantly associated with lymph node metastasis (P = 0.01), TNM staging (P = 0.003) and poor outcome of ESCC patients (P<0.05). Functional studies demonstrated that CACNA2D3 could inhibit tumorigenicity, cell motility and induce apoptosis. Mechanism study found that CACNA2D3 could arrest cell cycle at G1/S checkpoint by increasing expressions of p21 and p53 and decreasing expression of CDK2. In addition, CACNA2D3 could upregulate intracellular free cytosolic Ca(2+) and subsequently induce apoptosis. CONCLUSION: CACNA2D3 is a novel TSG responsible to the 3p21 deletion event and plays a critical suppressing role in the development and progression of ESCC.


Assuntos
Sequência de Bases , Canais de Cálcio/genética , Carcinoma de Células Escamosas/genética , Cromossomos Humanos Par 3 , Neoplasias Esofágicas/genética , Regulação Neoplásica da Expressão Gênica , Deleção de Sequência , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Cálcio/metabolismo , Canais de Cálcio/deficiência , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Pontos de Checagem do Ciclo Celular/genética , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...