Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Mol Med (Berl) ; 96(6): 547-558, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29748698

RESUMO

Elevated LRP16 expression is associated with poor clinical outcomes in multiple malignancies. We detected LRP16 expression in hepatocellular carcinoma (HCC) and found that it was downregulated in tumor samples and HCC cell lines. In a cohort of 80 HCC patients, high level of LRP16 expression in HCC tumors was associated with well differentiation, less lymph node metastasis, and good overall survival (OS). Overexpression of LRP16 in the HepG2 and MHCC-97L cell lines increased cell apoptosis, attenuated cell proliferation, migration, and invasion ability in vitro, and drastically diminished tumor growth and metastasis in vivo. Silencing LRP16 in HCC-LM3 and SMMC-7721 cell lines showed opposite results. Microarray evaluation of tumor cells overexpressing LRP16 revealed the effects on decreased activity in the Wnt signaling pathway. These results were confirmed by qRT-PCR and Western blots. Furthermore, inhibition of Wnt signaling decreased proliferation, migration, and invasion of HCC cell lines. Mechanism conducted showed that LRP16 overexpression could prevent ß-catenin from entering the nucleus. Our study demonstrated that LRP16 suppresses tumor growth in HCC by modulating Wnt/ß-catenin signaling. KEY MESSAGES: LRP16 was low expression in HCC tissue and cell lines. Low expression of LRP16 in HCC was associated with poor prognosis. LRP16 inhibits activation of the Wnt/ß-catenin pathway in HCC. LRP16 prevents ß-catenin from entering the nucleus.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Via de Sinalização Wnt , Idoso , Animais , Apoptose , Hidrolases de Éster Carboxílico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/genética , beta Catenina/metabolismo
2.
Onco Targets Ther ; 11: 1215-1222, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29551900

RESUMO

BACKGROUND: Leukemia related protein 16 (LRP16), one of the genes belonging to the macro domain family, has been found up-regulated in various tumors including testicles, ovaries and mucosa of colon and is associated with poor clinical outcomes. PURPOSE: The objective of this study was to investigate expression pattern and biological roles of LRP16 in pancreatic cancer. PATIENTS AND METHODS: Western blot and immunohistochemistry were used to investigate the expression of LRP16 in pancreatic cancer cell lines and tissues. qRT-PCR was utilized to examine LRP16 mRNA expression. Lentivirus based overexpression and knockdown of LRP16 was carried out in four pancreatic cancer cell lines (Panc1, CFPAC1, SW1990 and AsPC1). Cell proliferation, migration and invasion were determined by MTS and transwell assay, respectively. Flow cytometry was performed to investigate cell apoptosis. In vivo, the tumorigenic ability of LRP16 was determined in a NOD/SCID mouse model. RESULTS: In the present study, we found that LRP16 expression was increased in pancreatic tumor samples, compared with normal tissues. Moreover, the LRP16 expression was positive in 60.9% of 156 specimens and correlated with tumor size, clinical stage, distant metastasis and tumor differentiation. Multivariate Cox regression analysis revealed that the level of LRP16 expression was an independent prognostic factor for overall survival in pancreatic cancer patients. Furthermore, silencing of LRP16 significantly accelerated apoptosis, decrease proliferation, migration and invasion of pancreatic cancer cell lines in vitro. In contrast, overexpression of LRP16 attenuated apoptosis, promoted proliferation, migration and invasion. In addition, in vivo study revealed that down regulation of LRP16 could attenuate tumor growth and prolong the survival. On the contrary, up-regulation of LRP16 could promote tumor growth and shorten their survival. CONCLUSION: These findings suggest that LRP16 played an oncogenic role in pancreatic carcinoma.

3.
Biochem Biophys Res Commun ; 493(4): 1478-1484, 2017 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-28986261

RESUMO

As an extracellular matrix protein, osteopontin (OPN) has been shown to play an important role in regulation of the immune response to tumors. Myeloid-derived suppressor cells (MDSCs), a heterogeneous population of myeloid progenitors, are major components of the immune suppressive tumor microenvironment and contribute to tumor evasion of the immune response. However, the specific regulating mechanisms underlying MDSCs expansion remain unclear. Here, we found that MDSCs accumulated in the spleen and tumors of 3LL tumor-bearing mice. Supernatant collected from 3LL cells was able to induce the expansion of MDSCs in peripheral blood mononuclear cell (PBMC) in vitro. Results of enzyme linked immunosorbent assay showed high levels of OPN and matrix metalloproteinase-9 (MMP-9) in this supernatant. Silencing OPN can effectively reduce MDSCs frequency in vivo and in vitro. Furthermore, a specific fragment of OPN, OPN-32 kDa cleaved by MMP-9 was detected in the supernatant from 3LL cells. Overexpression of OPN-32 kDa in 3LL cells induced MDSCs expansion. Inhibition of MMP-9 by monoclonal antibody and inhibitor (TIMP-1) reduced MDSCs expansion in vitro and in vivo. These findings suggest that the MMP-9-cleaved OPN fragment, OPN-32kDa, was responsible for MDSCs expansion, which may contribute to tumor's evasion of the immune response.


Assuntos
Carcinoma Pulmonar de Lewis/enzimologia , Carcinoma Pulmonar de Lewis/imunologia , Metaloproteinase 9 da Matriz/metabolismo , Células Supressoras Mieloides/imunologia , Osteopontina/imunologia , Osteopontina/metabolismo , Evasão Tumoral/imunologia , Evasão Tumoral/fisiologia , Animais , Carcinoma Pulmonar de Lewis/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Supressoras Mieloides/patologia , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Microambiente Tumoral/imunologia , Microambiente Tumoral/fisiologia
4.
Int J Nanomedicine ; 12: 4019-4035, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28603416

RESUMO

The Chinese traditional medicine Shikonin is an ideal drug due to its multiple targets to tumor cells. But in clinics, improving its aqueous solubility and tumor accumulation is still a challenge. Herein, a copolymer with tunable poly(N-isopropylacrymaide) and polylactic acid block lengths is designed, synthesized, and characterized in nuclear magnetic resonance. The corresponding thermosensitive nanomicelle (TN) with well-defined core-shell structure is then assembled in an aqueous solution. For promoting the therapeutic index, the physical-chemistry properties of TNs including narrow size, low critical micellar concentration, high serum stability, tunable volume phase transition temperature (VPTT), high drug-loading capacity, and temperature-controlled drug release are systematically investigated and regulated through the fine self-assembly. The shikonin is then entrapped in a degradable inner core resulting in a shikonin-loaded thermosensitive nanomicelle (STN) with a VPTT of ~40°C. Compared with small-molecular shikonin, the in vitro cellular internalization and cytotoxicity of STN against breast cancer cells (Michigan Cancer Foundation-7) are obviously enhanced. In addition, the therapeutic effect is further enhanced by the programmed cell death (PCD) specifically evoked by shikonin. Interestingly, both the proliferation inhibition and PCD are synergistically promoted as T > VPTT, namely the temperature-regulated passive targeting. Consequently, as intravenous injection is administered to the BALB/c nude mice bearing breast cancer, the intratumor accumulation of STNs is significantly increased as T > VPTT, which is regulated by the in-house developed heating device. The in vivo antitumor assays against breast cancer further confirm the synergistically enhanced therapeutic efficiency. The findings of this study indicate that STN is a potential effective nanoformulation in clinical cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/terapia , Hipertermia Induzida/métodos , Naftoquinonas/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Neoplasias da Mama/tratamento farmacológico , Preparações de Ação Retardada , Medicamentos de Ervas Chinesas/química , Feminino , Humanos , Células MCF-7 , Camundongos Endogâmicos BALB C , Micelas , Nanoestruturas/química , Naftoquinonas/química , Naftoquinonas/farmacocinética , Poliésteres/química , Polímeros/química , Solubilidade , Temperatura , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
5.
World J Gastroenterol ; 22(26): 6036-48, 2016 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-27468195

RESUMO

AIM: To evaluate the efficacy of umbilical cord-derived mesenchymal stem cells (UC-MSCs) transplantation in the treatment of liver fibrosis. METHODS: Cultured human UC-MSCs were isolated and transfused into rats with liver fibrosis induced by dimethylnitrosamine (DMN). The effects of UC-MSCs transfusion on liver fibrosis were then evaluated by histopathology; serum interleukin (IL)-4 and IL-10 levels were also measured. Furthermore, Kupffer cells (KCs) in fibrotic livers were isolated and cultured to analyze their phenotype. Moreover, UC-MSCs were co-cultured with KCs in vitro to assess the effects of UC-MSCs on KCs' phenotype, and IL-4 and IL-10 levels were measured in cell culture supernatants. Finally, UC-MSCs and KCs were cultured in the presence of IL-4 antibodies to block the effects of this cytokine, followed by phenotypical analysis of KCs. RESULTS: UC-MSCs transfused into rats were recruited by the injured liver and alleviated liver fibrosis, increasing serum IL-4 and IL-10 levels. Interestingly, UC-MSCs promoted mobilization of KCs not only in fibrotic livers, but also in vitro. Co-culture of UC-MSCs with KCs resulted in increased production of IL-4 and IL-10. The addition of IL-4 antibodies into the co-culture system resulted in decreased KC mobilization. CONCLUSION: UC-MSCs could increase IL-4 and promote mobilization of KCs both in vitro and in vivo, subsequently alleviating the liver fibrosis induced by DMN.


Assuntos
Interleucina-10/imunologia , Interleucina-4/imunologia , Células de Kupffer/imunologia , Cirrose Hepática/terapia , Fígado/imunologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Cordão Umbilical/citologia , Animais , Movimento Celular , Células Cultivadas , Técnicas de Cocultura , Dimetilnitrosamina/toxicidade , Modelos Animais de Doenças , Humanos , Técnicas In Vitro , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Ratos , Ratos Sprague-Dawley
6.
Int J Clin Exp Med ; 8(8): 12765-73, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26550190

RESUMO

BACKGROUNDS: Non-small cell lung cancer (NSCLC) is one of the most common malignancies with a high mortality level. Recently, a variety of studies explored the role of osteopontin (OPN) expression in the prognosis of NSCLC, but the results were controversial. METHODS: We performed a meta-analysis of eligible studies to evaluate the prognostic significance of OPN expression in NSCLC patients. In order to assess the association between OPN and OS and DFS/PFS, hazard ratio (HR) with 95% confidence interval (CI) was calculated. RESULTS: A total of ten studies comprising 1420 patients were included in the meta-analysis. The summary results indicated that high OPN expression was a poor predictor for OS (HR = 2.19, 95% CI: 1.6-2.98), and DFS/PFS (HR = 2, 95% CI: 1.66-2.41). Subgroup analysis revealed that high OPN expression was a negative prognostic marker for OS and DFS/PFS regardless of ethnicity background, treatment and OPN detection method. CONCLUSION: Our results showed that increased OPN expression significantly correlated with poor OS and DPS/PFS in NSCLC patients.

7.
Int J Biol Markers ; 30(4): e359-63, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26165685

RESUMO

OBJECTIVE: To explore the prognostic value of serum interleukin-17 (IL-17) and vascular endothelial growth factor (VEGF) levels in patients with small cell lung cancer (SCLC). MATERIALS AND METHODS: IL-17 and VEGF levels were determined by a commercially available ELISA method. RESULTS: Serum IL-17 and VEGF levels were higher in the SCLC group than the control group (p<0.001 and p<0.0001, respectively). In addition, there was a significant correlation between IL-17 and VEGF. The level of IL-17 showed significant correlations with tumor stage and tumor metastasis, while serum VEGF levels were significantly associated only with tumor metastasis. Univariate and multivariate analysis indicated that an elevated IL-17 level was an independent prognostic factor for shorter overall survival in SCLC. CONCLUSIONS: IL-17 may play an important role in tumor dissemination in SCLC patients and measurement of IL-17 could have useful prognostic value for worse overall survival in patients with SCLC.


Assuntos
Biomarcadores Tumorais/sangue , Interleucina-17/sangue , Neoplasias Pulmonares/sangue , Carcinoma de Pequenas Células do Pulmão/sangue , Fator A de Crescimento do Endotélio Vascular/sangue , Estudos de Casos e Controles , Feminino , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prognóstico , Modelos de Riscos Proporcionais , Estudos Prospectivos , Carcinoma de Pequenas Células do Pulmão/mortalidade , Carcinoma de Pequenas Células do Pulmão/secundário
8.
Biochem Biophys Res Commun ; 464(1): 249-55, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26111447

RESUMO

Thymosin α1 (Tα1) has been tested for cancer therapy for several years, in most cases, the anti-tumor effect of Tα1 was limited, especially when Tα1 was used as a single agent. The role of Tα1 in cancer treatment and the regulatory mechanisms by which Ta1 takes effects are not yet completely understood. Using a Lewis lung caner model, here we report that Tα1 used alone elevated CD8(+) T cells, but failed to inhibit tumor growth. Furthermore, immunosuppressive myeloid-derived suppressor cells (MDSCs) showed heightened Arginase 1 production in response to Tα1 treatment, which led to stronger suppression of anti-tumor immunity. In addition, the upregulation of ARG1 was dependent on TLRs/MyD88 signaling, blocking MyD88 signaling abrogated the enhanced ARG1 expression and restored the anti-tumor efficacy of Tα1. This study provides the first demonstration that Tα1 treatment activates but not expands MDSCs via MyD88 signaling, which indicates better immunotherapeutic strategy of Tα1 against cancer.


Assuntos
Antineoplásicos/farmacologia , Arginase/imunologia , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , Células Mieloides/efeitos dos fármacos , Timosina/análogos & derivados , Animais , Arginase/genética , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/mortalidade , Ativação Enzimática/efeitos dos fármacos , Feminino , Imunidade Inata/efeitos dos fármacos , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/patologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais , Análise de Sobrevida , Timalfasina , Timosina/farmacologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
9.
Cancer Biomark ; 15(4): 425-32, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25792471

RESUMO

BACKGROUND: High expression of CD14(+)HLA-DR-/low myeloid-derived suppressor cells (MDSCs) is correlated with immunosuppressive activity in various cancers, however, no studies have shown a correlation of these immunosuppressive cells with clinical outcomes in small-cell lung cancer (SCLC) patients. OBJECTIVE: The purpose of the study was to investigate the number, frequency and clinical significance of CD14(+)HLA-DR-/low MDSCs in SCLC patients. METHODS: The peripheral blood of 42 patients with SCLC and 37 healthy controls was analyzed by using flow cytometry. The relationships between the number and frequency of MDSCs, clinicopathological features and overall survival(OS) were analyzed. The prognostic value of MDSCs was tested by using univariate and multivariate analysis. RESULTS: Our results demonstrated that number and frequency of peripheral CD14(+)HLA-DR-/low MDSCs were significantly increased in SCLC patients than in controls (p< 0.0001 and p< 0.0001, respectively). The frequency of MDSCs correlated with tumor stage (p= 0.02), serum LDH levels (p= 0.001) and with the poorer OS (log-rank test, p= 0.017). Univariate and multivariate analyses suggested that frequency of CD14(+)HLA-DR-/low MDSCs as an independent biomarker for poor prognosis in SCLC patients during follow-up. Our study provides the first evidence that the frequency of CD14(+)HLA-DR-/low MDSCs negatively correlates with clinical outcomes in SCLC patients. CONCLUSIONS: The frequency of CD14(+)HLA-DR-/low MDSCs could be considered as a poor prognostic predictor in SCLC and the elimination of MDSCs during medical interventions may improve the prognosis of SCLC patients.


Assuntos
Biomarcadores Tumorais/sangue , Antígenos HLA-DR/imunologia , Receptores de Lipopolissacarídeos/imunologia , Carcinoma de Pequenas Células do Pulmão/imunologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/imunologia , Feminino , Citometria de Fluxo , Antígenos HLA-DR/genética , Humanos , Receptores de Lipopolissacarídeos/genética , Masculino , Pessoa de Meia-Idade , Células Mieloides/imunologia , Células Mieloides/patologia , Prognóstico , Carcinoma de Pequenas Células do Pulmão/sangue , Carcinoma de Pequenas Células do Pulmão/genética
10.
Tumour Biol ; 36(4): 2651-6, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25480413

RESUMO

Interleukin-35 (IL-35) has recently been implicated in tumor immunity. The aim of this study was to explore the clinical role of plasma IL-35 in patients with non-small cell lung cancer (NSCLC). Plasma collected from 106 patients with NSCLC cases and 78 healthy controls (HC) were subjected to IL-35 enzyme-linked immunosorbent assay (ELISA) and relationships between plasma IL-35 levels and clinical characteristics were evaluated. The correlation of IL-35 and overall survival was analyzed using Kaplan-Meier method. The prognostic value of IL-35 was tested using univariate and multivariate analysis. Circulating IL-35 levels were significantly higher in the NSCLC group in comparison with the HC group (21.37 ± 11.55 pg/ml vs. 10.09 ± 5.32 pg/ml, p < 0.001). Correlation analysis by subgroup indicated that plasma IL-35 correlated positively with tumor TNM stage (p < 0.001) and lymph node metastases (p < 0.0001). Using a cutoff level of 20.26 pg/ml (median value), IL-35 showed an inverse correlation with overall survival. Univariate and multivariate analysis indicated that plasma IL-35 was an independent prognostic factor for NSCLC patients. Circulating IL-35 in NSCLC patients significantly increased. IL-35 is a promising potential biomarker in prognostication of clinical outcome of NSCLC patients and the regulation of IL-35 expression may provide a new target for the treatment of NSCLC patients.


Assuntos
Biomarcadores Tumorais/sangue , Carcinoma Pulmonar de Células não Pequenas/sangue , Interleucinas/sangue , Prognóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/patologia , Intervalo Livre de Doença , Feminino , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade
11.
Oncotarget ; 5(18): 8317-29, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25327561

RESUMO

Although HER2-targeting antibody trastuzumab confers a substantial benefit for patients with HER2-overexpressing breast and gastric cancer, overcoming trastuzumab resistance remains a large unmet need. In this study, we revealed a STAT3-centered positive feedback loop that mediates the resistance of trastuzumab. Mechanistically, chronic exposure of trastuzumab causes the upregulation of fibronection (FN), EGF and IL-6 in parental trastuzumab-sensitive breast and gastric cells and convergently leads to STAT3 hyperactivation. Activated STAT3 enhances the expression of FN, EGF and IL-6, thus constituting a positive feedback loop which amplifies and maintains the STAT3 signal; furthermore, hyperactivated STAT3 signal promotes the expression of MUC1 and MUC4, consequently mediating trastuzumab resistance via maintenance of persistent HER2 activation and masking of trastuzumab binding to HER2 respectively. Genetic or pharmacological inhibition of STAT3 disrupted STAT3-dependent positive feedback loop and recovered the trastuzumab sensitivity partially due to increased apoptosis induction. Combined trastuzumab with STAT3 inhibition synergistically suppressed the growth of the trastuzumab-resistant tumor xenografts in vivo. Taken together, our results suggest that feedback activation of STAT3 constitutes a key node mediating trastuzumab resistance. Combinatorial targeting on both HER2 and STAT3 may enhance the efficacy of trastuzumab or other HER2-targeting agents in HER2-positive breast and gastric cancer.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Mucina-1/metabolismo , Mucina-4/metabolismo , Fator de Transcrição STAT3/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Ácidos Aminossalicílicos/farmacologia , Animais , Benzenossulfonatos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fator de Crescimento Epidérmico/metabolismo , Retroalimentação Fisiológica , Feminino , Fibronectinas/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Interleucina-6/metabolismo , Camundongos Nus , Mucina-1/genética , Mucina-4/genética , Fosforilação , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fatores de Tempo , Trastuzumab , Carga Tumoral/efeitos dos fármacos , Regulação para Cima
12.
J Immunol ; 193(11): 5461-9, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25344470

RESUMO

Although myeloid-derived suppressor cells (MDSCs) are well known for their immunosuppressive function in several pathological conditions, the role of MDSCs in hepatitis B virus infection remains obscure. In this study, we investigated the frequency and function of MDSCs in the peripheral blood and liver of 91 chronic hepatitis B (CHB) patients. A higher percentage of MDSCs, defined as CD14(+)HLA-DR(-/low), was detected in peripheral blood of CHB patients than that of the healthy controls. Moreover, high expression of programmed death 1 (PD-1) and secretion of IL-10 in this population were determined. The frequency of MDSCs was positively correlated with serum viral load, but it was negatively correlated with liver inflammatory injury. These cells were also abundant in liver tissue of CHB patients and were related to necroinflammatory activity. Furthermore, we found that these cells could suppress hepatitis B virus-specific CD8(+) T cell response, including reduced proliferation and IFN-γ production, and inhibit degranulation of CD8(+) T cells, including reduced production of granzyme B and perforin. Importantly, PD-1-induced IL-10 production by MDSCs was responsible for the suppressive activity. To our knowledge, for the first time our study proved that CD14(+)HLA-DR(-/low)PD-1(+) MDSCs in CHB patients contribute to an inadequate immune response against the virus and lead to chronic infection, which represents a potential target for therapeutic intervention.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Vírus da Hepatite B/imunologia , Hepatite B Crônica/imunologia , Células Mieloides/imunologia , Adulto , Linfócitos T CD8-Positivos/virologia , Degranulação Celular , Células Cultivadas , Feminino , Humanos , Terapia de Imunossupressão , Interleucina-10/genética , Interleucina-10/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Pessoa de Meia-Idade , Células Mieloides/virologia , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Regulação para Cima , Adulto Jovem
13.
Biochem Biophys Res Commun ; 452(3): 795-800, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25201732

RESUMO

Osteopontin (OPN) is abundant in mineralized tissues and has long been implicated in bone remodeling. However, the therapeutic effect of targeting OPN in bone loss diseases and the underlying molecular mechanism remain largely unknown. Here, we reported that anti-OPN mAb (23C3) could protect against ovariectomy-induced osteoporosis in mice, demonstrated by microcomputed tomography analysis and histopathology evaluation. In vitro assay showed that 23C3 mAb reduced osteoclasts (OCs)-mediated bone resorption through promotion of mature OC apoptosis. Thus, the study has important implications for understanding the role of OPN in OC bone resorption and survival, and OPN antagonists may have therapeutic potential for osteoporosis and other osteopenic diseases.


Assuntos
Anticorpos Monoclonais/farmacologia , Reabsorção Óssea/tratamento farmacológico , Osteoclastos/efeitos dos fármacos , Osteopontina/antagonistas & inibidores , Osteoporose/tratamento farmacológico , Ovariectomia/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/etiologia , Reabsorção Óssea/genética , Células Cultivadas , Feminino , Expressão Gênica , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteopontina/genética , Osteopontina/metabolismo , Osteoporose/diagnóstico por imagem , Osteoporose/etiologia , Osteoporose/genética , Microtomografia por Raio-X
14.
Mol Cancer Ther ; 13(8): 2127-37, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24928851

RESUMO

Mesenchymal stem cells (MSC) represent a new tool for delivery of therapeutic agents to cancer sites because of their strong tropism toward tumors. IL15 has demonstrated a potent antitumor activity in various animal models as well as clinical trials. However, because of its short half-life, effective therapeutic effects usually require a high dose, which often results in undesired side effects; thus, new strategies for overcoming this disadvantage are needed. In this study, human MSCs were isolated from umbilical cord blood as delivery vehicles and transduced with lentivirus vector expressing murine IL15 (MSC-IL15). In vitro assays of lymphocyte activation and proliferation demonstrated that IL15 produced by MSCs was biofunctional. In syngeneic mice bearing Pan02 pancreatic tumors, systemic administration of MSC-IL15 significantly inhibited tumor growth and prolonged the survival of tumor-bearing mice, which were associated with tumor cell apoptosis, and natural killer (NK)- and T-cell accumulation. Furthermore, we confirmed that MSC-IL15 could migrate toward tumor and secreted IL15 in tumor-specific sites. Depletion of NK and CD8(+) T cells abolished the antitumor activity of MSC-IL15, suggesting that NK and CD8(+) T cells play a key role for MSC-IL15-mediated effect. Interestingly, cured mice after MSC-IL15 treatment were resistant to Pan02 pancreatic tumor rechallenge, and adoptive transfer of lymphocytes from cured mice also could cause rejection of Pan02 tumor inoculation in naïve mice, indicating that MSC-IL15 induced tumor-specific T-cell immune memory response. Overall, these data support that MSCs producing IL15 might represent an innovative strategy for therapy of pancreatic tumor.


Assuntos
Carcinoma Ductal Pancreático/terapia , Transplante de Células-Tronco Mesenquimais , Neoplasias Pancreáticas/terapia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/imunologia , Carcinoma Ductal Pancreático/imunologia , Linhagem Celular Tumoral , Movimento Celular , Sangue Fetal/citologia , Humanos , Interleucina-15/metabolismo , Células Matadoras Naturais/imunologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Pancreáticas/imunologia
15.
PLoS One ; 8(12): e84927, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24367702

RESUMO

There is an urgent need for improved therapy for advanced ovarian carcinoma, which may be met by administering immune-modulatory monoclonal antibodies (mAbs) to generate a tumor-destructive immune response. Using the ID8 mouse ovarian cancer model, we investigated the therapeutic efficacy of various mAb combinations in mice with intraperitoneal (i.p.) tumor established by transplanting 3 × 10(6) ID8 cells 10 days previously. While most of the tested mAbs were ineffective when given individually or together, the data confirm our previous finding that 2 i.p. injections of a combination of anti-CD137 with anti-PD-1 mAbs doubles overall survival. Mice treated with this mAb combination have a significantly increased frequency and total number of CD8(+) T cells both in the peritoneal lavage and spleens, and these cells are functional as demonstrated by antigen-specific cytolytic activity and IFN-γ production. While administration of anti-CD137 mAb as a single agent similarly increases CD8(+) T cells, these have no functional activity, which may be attributed to up-regulation of co-inhibitory PD-1 and TIM-3 molecules induced by CD137. Addition of the anti-cancer drug cisplatin to the 2 mAb combination increased overall survival >90 days (and was probably curative) by a mechanism which included a systemic CD8(+) T cell response with tumor specificity and immunological memory. Strikingly, combined treatment of cisplatin and CD137/PD-1 mAb also gave rise to the long-term survival of mice with established TC1 lung tumors. A similar combination of the 2 mAbs and cisplatin should be considered for clinical 'translation'.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/imunologia , Cisplatino/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Análise de Variância , Animais , Anticorpos Monoclonais Murinos/administração & dosagem , Anticorpos Monoclonais Murinos/uso terapêutico , Sinergismo Farmacológico , Ensaio de Imunoadsorção Enzimática , Feminino , Injeções Intraperitoneais , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/administração & dosagem , Receptor de Morte Celular Programada 1/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/administração & dosagem , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
16.
Clin Cancer Res ; 19(17): 4697-705, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23857601

RESUMO

PURPOSE: Giant cell tumors of bone (GCTB) exhibit aggressive bone lytic behavior. Studies have shown that interleukin 17A (IL-17A) is involved pathologic bone resorption in various skeletal disorders. Thus, we have investigated the role of IL-17A in GCTBs. EXPERIMENTAL DESIGN: We evaluated the progression of GCTBs using Campanacci grading and Enneking staging systems in 74 patients with GCTB. The expression of IL-17A and the IL-17A receptor A (IL-17RA) was assessed in GCTB tissues and in both multinucleated giant cells (MNGC) and stromal cells cultured in vitro using immunostaining and reverse transcription PCR (RT-PCR). The effects of IL-17A on the osteolytic activity of the MNGCs and the proliferation of the stromal cells were investigated using the "pit" formation and MTT assays, respectively. The effects of IL-17A on the expression of proosteolytic factors were examined in primary cultured MNGCs and stromal cells using RT-PCR, Western blotting, and gene expression microarrays. RESULTS: In GCTBs, we detected abundant levels of IL-17A, which were associated with tumor extension and grade. IL-17A is predominantly produced by MNGCs, whereas IL-17RA is expressed by both MNGCs and stromal cells in GCTBs. In the MNGCs, the IL-17A increased the mRNA expression of IL-17A and proosteolytic enzymes, and also enhanced osteolytic ability. In the stromal cells, the IL-17A stimulated cellular proliferation and the expression of proosteolytic factors, including RANKL through myc and STAT3, respectively. In addition, IL-17A stimulated in vivo tumor growth and the extent of angiogenesis in GCTBs. CONCLUSION: IL-17A stimulates the progression of GCTBs and might represent a useful candidate marker for progression and as a therapeutic target for GCTBs.


Assuntos
Neoplasias Ósseas/genética , Carcinogênese/genética , Tumor de Células Gigantes do Osso/genética , Interleucina-17/biossíntese , Adulto , Idoso , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Proliferação de Células , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Tumor de Células Gigantes do Osso/patologia , Humanos , Interleucina-17/genética , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais/genética
17.
Cancer Immunol Immunother ; 62(9): 1439-51, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23760662

RESUMO

Accumulating evidence has demonstrated that myeloid-derived suppressor cells (MDSCs), a heterogeneous population of cells, play an important role in the subversion, inhibition, and downregulation of the immune response to cancer. However, the characteristics of these cells, particularly clinical relevance, in malignant tumors remain unclear due to a lack of specific markers. In this study, we characterized peripheral CD14(+)HLA-DR(-/low) cells, a new human MDSC subpopulation, in 89 patients with non-small cell lung cancer (NSCLC). As expected, both frequency and absolute number of CD14(+)HLA-DR(-/low) cells were significantly increased in the peripheral blood of NSCLC patients compared with that of the healthy controls and indicated an association with metastasis, response to chemotherapy, and progression-free survival. These cells showed decreased expression of CD16 and CD86 compared with HLA-DR(+) monocytes. Unlike classical monocytes, these populations showed significantly decreased allostimulatory activity and showed the ability to inhibit autologous T cell proliferation and IFN-γ production in a cell-contact-dependent manner. Furthermore, we demonstrated that CD14(+)HLA-DR(-/low) cells expressed the NADPH oxidase component gp91(phox) and generated high level of reactive oxygen species (ROS). Moreover, inactivation of ROS reversed their immunosuppressive capacity on T cell response. These results prove, for the first time, the existence of ROS-producing CD14(+)HLA-DR(-/low) myeloid-derived suppressor cells in NSCLC patients, which mediate tumor immunosuppression and might thus represent a potential target for therapeutic intervention.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Antígenos HLA-DR/imunologia , Receptores de Lipopolissacarídeos/imunologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Células Mieloides/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/patologia , Processos de Crescimento Celular/imunologia , Cisplatino/administração & dosagem , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Intervalo Livre de Doença , Docetaxel , Feminino , Citometria de Fluxo , Glutamatos/administração & dosagem , Guanina/administração & dosagem , Guanina/análogos & derivados , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Células Mieloides/patologia , Pemetrexede , Taxoides/administração & dosagem , Gencitabina
18.
Mol Immunol ; 54(1): 74-83, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23220070

RESUMO

Cimetidine, a histamine type-2 receptor antagonist, is known to inhibit the growth of several tumors in human and animals, however the mechanism of action underlying this effect remains largely unknown. Here, in the mice model of 3LL lung tumor, cimetidine showed significant inhibition of tumor growth. However, an in vitro study demonstrated that cimetidine showed no effect on proliferation, survival, migration and invasion of 3LL cells. We found that cimetidine reduced CD11b(+)Gr-1(+) myeloid derived-suppressive cell (MDSC) accumulation in spleen, blood and tumor tissue of tumor-bearing mice. In vitro coculture assay showed that cimetidine reversed MDSC-mediated T-cell suppression, and improved IFN-γ production. Further investigation demonstrated that the NO production and arginase I expression of MDSCs were reduced, and MDSCs prone to apoptosis by cimetidine treatment. However, MDSC differentiation was not affect by cimetidine. Importantly, although histamine H2 receptor was expressed in MDSC surface, histamine could not reverse the proapoptosis of cimetidine. Moreover, famotidine also did not have this capacity. We found that cimetidine could induce Fas and FasL expression in MDSC surface, and sequentially regulate caspase-dependent apoptosis pathway. Thus, these findings revealed a novel mechanism for cimetidine to inhibit tumor via modulation of MDSC apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Lewis/patologia , Proliferação de Células/efeitos dos fármacos , Cimetidina/farmacologia , Neoplasias Pulmonares/patologia , Células Mieloides/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Antagonistas dos Receptores H2 da Histamina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Biológicos , Células Mieloides/patologia , Células Mieloides/fisiologia , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/patologia , Células Progenitoras Mieloides/fisiologia
19.
PLoS One ; 7(12): e50850, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251393

RESUMO

It is recognized that endogenous cannabinoids, which signal through CB1 receptors in hepatic stellate cells (HSCs), exert a profibrotic effect on chronic liver diseases. In this study, we suppressed CB1 expression by lentivirus mediated small interfering RNA (CB1-RNAi-LV) and investigated its effect on hepatic fibrosis in vitro and in vivo. Our results demonstrated that CB1-RNAi-LV significantly inhibited CB1 expression, and suppressed proliferation and extracellular matrix production in HSCs. Furthermore, CB1-RNAi-LV ameliorated dimethylnitrosamine induced hepatic fibrosis markedly, which was associated with the decreased expression of mesenchymal cell markers smooth muscle α-actin, vimentin and snail, and the increased expression of epithelial cell marker E-cadherin. The mechanism lies on the blockage of Smad signaling transduction induced by transforming growth factor ß1 and its receptor TGF-ß RII. Our study firstly provides the evidence that CB1-RNAi-LV might ameliorate hepatic fibrosis through the reversal of epithelial-to-mesenchymal transition (EMT), while the CB1 antagonists AM251 had no effect on epithelial-mesenchymal transitions of HSCs. This suggests that CB1 is implicated in hepatic fibrosis and selective suppression of CB1 by small interfering RNA may present a powerful tool for hepatic fibrosis treatment.


Assuntos
Transição Epitelial-Mesenquimal/genética , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/genética , Fígado/metabolismo , Receptor CB1 de Canabinoide/genética , Animais , Apoptose/genética , Caderinas/genética , Caderinas/metabolismo , Proliferação de Células , Células Estreladas do Fígado/patologia , Fígado/patologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/metabolismo
20.
Mol Immunol ; 48(15-16): 2019-26, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21737140

RESUMO

Osteopontin (OPN) is a cytokine with multiple functions, including the regulation of innate immune response. However, the detailed function and mechanism of OPN in host defense against invaded microorganisms remain unclear. In this report, we revealed that OPN could affect the clearance of Propionibacterium acnes in kupffer cells. In a murine model of P. acnes induced hepatic granuloma, OPN-deficient mice or wild-type (WT) mice treated with anti-OPN mAb exhibited more hepatic granuloma formation than WT mice. Increased infiltration of intrahepatic leukocytes, higher expression of TLRs, and significantly upregulated level of proinflammatory cytokines of liver tissue were observed in OPN-deficient mice after P. acnes challenge. Moreover, in vitro assay showed that kupffer cells isolated from OPN(-/-) mice exhibited impairment in clearance of P. acnes. Kupffer cells isolated from OPN(-/-) mice showed reduced level of NADPH oxidase-mediated reactive oxygen species (ROS) in response to P. acnes, which was regulated by NADPH oxidase subunit p47phox. Further investigation revealed that OPN interaction with αvß3 integrin activated PI3K and ERK signal pathways, leading to the expression of p47phox. Taken together, these data demonstrated an important role of OPN in enhancing the antimicrobial innate immune response by modulation of bacterium clearance activity in kupffer cells.


Assuntos
Regulação da Expressão Gênica/imunologia , Infecções por Bactérias Gram-Positivas/imunologia , Células de Kupffer/imunologia , NADPH Oxidases/biossíntese , Osteopontina/imunologia , Animais , Western Blotting , Separação Celular , Citometria de Fluxo , Expressão Gênica , Infecções por Bactérias Gram-Positivas/metabolismo , Infecções por Bactérias Gram-Positivas/patologia , Granuloma/imunologia , Granuloma/microbiologia , Granuloma/patologia , Células de Kupffer/metabolismo , Fígado/imunologia , Fígado/microbiologia , Fígado/patologia , Camundongos , Camundongos Knockout , NADPH Oxidases/imunologia , Osteopontina/metabolismo , Propionibacterium acnes/imunologia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA