Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 34(5): 729-40, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21530327

RESUMO

CD28 and CTLA-4 are cell surface cosignaling molecules essential for the control of T cell activation upon the engagement of their ligands B7-1 and B7-2 from antigen-presenting cells. By employing a receptor array assay, we have demonstrated that B7-H2, best known as the ligand of inducible costimulator, was a ligand for CD28 and CTLA-4 in human, whereas these interactions were not conserved in mouse. B7-H2 and B7-1 or B7-2 interacted with CD28 through distinctive domains. B7-H2-CD28 interaction was essential for the costimulation of human T cells' primary responses to allogeneic antigens and memory recall responses. Similar to B7-1 and B7-2, B7-H2 costimulation via CD28 induced survival factor Bcl-xL, downregulated cell cycle inhibitor p27(kip1), and triggered signaling cascade of ERK and AKT kinase-dependent pathways. Our findings warrant re-evaluation of CD28 and CTLA-4's functions previously attributed exclusively to B7-1 and B7-2 and have important implications in therapeutic interventions against human diseases.


Assuntos
Antígenos CD/imunologia , Antígenos CD28/imunologia , Animais , Antígenos CD/química , Antígenos de Diferenciação de Linfócitos T/imunologia , Sítios de Ligação , Antígenos CD28/química , Antígeno CTLA-4 , Linhagem Celular , Proliferação de Células , Humanos , Ligante Coestimulador de Linfócitos T Induzíveis , Proteína Coestimuladora de Linfócitos T Induzíveis , Ligantes , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Estrutura Quaternária de Proteína , Linfócitos T/citologia , Linfócitos T/imunologia
2.
Hepatology ; 68(6): 2239-2253, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29729199

RESUMO

Polarity defects are frequently involved in liver diseases, such as chronic hepatitis and hepatocellular carcinoma (HCC). It was reported that vacuolar protein sorting 33B (Vps33b) plays critical roles in the maintenance of hepatocyte polarity; however, the functional roles and mechanisms of Vps33b in HCC occurrence and progression remain unknown. First of all, we showed that Vps33b is down-regulated in human and mouse liver cancer samples, and the low expression levels of Vps33b correlate with the poor prognosis of many HCC patients. Liver-specific Vps33b deficiency induces liver damage, progressive hepatitis, fibrosis, and HCC in male mice, indicating that Vps33b is a crucial contributory factor to hepatocarcinogenesis. Vps33b deficiency-caused liver damage was primarily due to the disorders of structural and functional hepatocyte polarity, which were reflected by the decreased protein levels of E-cadherin because of inaccurate location to lysosomes and polarity defects at both apical and lateral plasma membrane proteins. The results of a mechanism study revealed that Vps33b interacts with VPS33B-interacting protein, which is involved in polarity and apical protein restriction; vesicle-trafficking protein Sec22b; and Flotillin-1 in hepatocytes and is in charge of the normal distribution of polarity-determined proteins. Expression levels of Vps33b negatively correlated with the degree of inflammatory cell infiltration in livers from diethylnitrosamine-induced or transgenic HCC mouse models, and the inflammatory stimuli suppressed the expression of Vps33b in vitro. Conclusion: Down-regulation of Vps33b expression is a critical step for inflammation-driven HCC, and Vps33b serves as an important tumor suppressor in hepatocarcinogenesis.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Caderinas/metabolismo , Carcinoma Hepatocelular/mortalidade , Membrana Celular/ultraestrutura , Progressão da Doença , Fibrose , Células Hep G2 , Hepatite/metabolismo , Hepatócitos/metabolismo , Humanos , Interleucina-6/metabolismo , Fígado/patologia , Neoplasias Hepáticas Experimentais/etiologia , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Proteínas R-SNARE/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas de Transporte Vesicular/genética
3.
Cancer Cell Int ; 19: 322, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31827396

RESUMO

BACKGROUND: Autologous tumor-infiltrating lymphocytes (Tils) immunotherapy is a promising treatment in patients with advanced hepatocellular cancer. Although Tils treatment has shown great promise, their persistence and the efficacy after adoptive-transfer are insufficient and remain a challenge. Studies have demonstrated that IL-15 and Akt inhibitor can regulate T cell differentiation and memory. Here, we constructed S-15 (Super human IL-15), a fusion protein consisting of human IL-15, the sushi domain of the IL-15 receptor α chain and human IgG-Fc. Herein we compared the effects of S-15 with IL-2 or in combination with Akti on the expansion and activation of Tils. METHODS: Hepatocellular cancer tissues were obtained from 6 patients, Tils were expanded using IL-2, IL-2/S-15, IL-2/Akti or in combination IL-2/S-15/Akti. At day 10, anti-CD3 antibody was added to the culture media and expanded to day 25. The composition, exhaustion and T-cell differentiation markers (CD45RA/CCR7) were analyzed by flow cytometry. RESULTS: We found that IL-2/S-15/Akti expanded Tils and showed the highest percentage of central memory CD45RA-CCR7+ phenotype prior to anti-CD3 antibody activation and after anti-CD3 antibody activation. T cells cultured with IL-2/S-15/Akti exhibited a mixture of CD4+, CD8+, and CD3+CD4-CD8- T cells; S-15 in combination with Akt inhibitor downregulated the expression of PD-1+Tim-3+ on Tils and decreased the Tregs in Tils. Additionally, the Tils expanded in the presence of the Akt inhibitor and S-15 showed enhanced antitumor activity as indicated by the increase in IFN-γ producing tumor infiltrating CD8+ T cells and without comprising the Tils expansion. CONCLUSION: Our study elucidates that IL-2/S-15/Akti expanded Tils and represent a viable source for the cellular therapy for patients with hepatocellular cancer.

4.
BMC Cancer ; 19(1): 279, 2019 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-30922248

RESUMO

BACKGROUND: With poor prognosis and limited treatment options for advanced hepatocellular carcinoma (HCC), development of novel therapeutic agents is urgently needed. This single-arm phase I study sought to assess the safety and preliminary efficacy of icaritin in human as a potential oral immunotherapy in addition to the immune-checkpoint inhibitors. METHODS: Eligible advanced HCC patients with Child-Pugh Class A or B were administered with a fixed oral dose of icaritin at either 600 or 800 mg b.i.d. The primary endpoint was safety, and the secondary endpoints included time-to-progression (TTP), overall survival (OS) and the clinical benefit rate (CBR). Icaritin treatment induced immune biomarkers and immune-modulating activities in myeloid cells were also explored. RESULTS: No drug-related adverse events ≥ Grade 3 were observed in all 20 enrolled HCC patients. Among the 15 evaluable patients, 7 (46.7%) achieved clinical benefit, representing one partial response (PR, 6.7%) and 6 stable disease (SD, 40%). The median TTP was 141 days (range: 20-343 days), and the median OS was 192 days (range: 33-1036 days). Durable survival was observed in PR/SD patients with a median OS of 488 days (range: 72-773). TTP was significantly associated with the dynamic changes of peripheral neutrophils (p = 0.0067) and lymphocytes (p = 0.0337). Icaritin treatment induced changes in immune biomarkers-and immune-suppressive myeloid cells were observed. CONCLUSIONS: Icaritin demonstrated safety profiles and preliminary durable survival benefits in advanced HCC patients, which were correlated with its immune-modulation activities and immune biomarkers. These results suggested the potential of icaritin as a novel oral immunotherapy for advanced HCC in addition to antibody-based PD-1/PD-L1 blockade therapies. TRIAL REGISTRATION: Clinicaltrial.gov identifier. NCT02496949 (retrospectively registered, July 14, 2015).


Assuntos
Biomarcadores Tumorais/imunologia , Carcinoma Hepatocelular/tratamento farmacológico , Flavonoides/administração & dosagem , Neoplasias Hepáticas/tratamento farmacológico , Administração Oral , Adulto , Idoso , Carcinoma Hepatocelular/imunologia , Esquema de Medicação , Feminino , Flavonoides/efeitos adversos , Flavonoides/farmacologia , Humanos , Neoplasias Hepáticas/imunologia , Linfócitos/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Neutrófilos/efeitos dos fármacos , Análise de Sobrevida , Resultado do Tratamento
5.
J Immunol ; 194(10): 4997-5006, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25876763

RESUMO

Tumor resistance is a major hurdle to anti-Her2/neu Ab-based cancer therapy. Current strategies to overcome tumor resistance focus on tumor cell-intrinsic resistance. However, the extrinsic mechanisms, especially the tumor microenvironment, also play important roles in modulating the therapeutic response and resistance of the Ab. In this study, we demonstrate that tumor progression is highly associated with TAMs with immune-suppressive M2 phenotypes, and deletion of TAMs markedly enhanced the therapeutic effects of anti-Her2/neu Ab in a HER2/neu-dependent breast cancer cell TUBO model. Tumor local delivery of IL-21 can skew TAM polarization away from the M2 phenotype to a tumor-inhibiting M1 phenotype, which rapidly stimulates T cell responses against tumor and dramatically promotes the therapeutic effect of anti-Her2 Ab. Skewing of TAM polarization by IL-21 relies substantially on direct action of IL-21 on TAMs rather than stimulation of T and NK cells. Thus, our findings identify the abundant TAMs as a major extrinsic barrier for anti-Her2/neu Ab therapy and present a novel approach to combat this extrinsic resistance by tumor local delivery of IL-21 to skew TAM polarization. This study offers a therapeutic strategy to modulate the tumor microenvironment to overcome tumor-extrinsic resistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos/imunologia , Interleucinas/imunologia , Macrófagos/imunologia , Neoplasias Mamárias Experimentais/imunologia , Microambiente Tumoral/imunologia , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Interleucinas/administração & dosagem , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real , Receptor ErbB-2/antagonistas & inibidores , Trastuzumab
7.
Adv Exp Med Biol ; 909: 1-67, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27240456

RESUMO

Antibody-based immunotherapy has become a standard treatment for a variety of cancers. Many well-developed antibodies disrupt signaling of various growth factor receptors for the treatment of a number of cancers by targeting surface antigens expressed on tumor cells. In recent years, a new family of antibodies is currently emerging in the clinic, which target immune cells rather than cancer cells. These immune-targeted therapies strive to augment antitumor immune responses by antagonizing immunosuppressive pathways or providing exogenous immune-activating stimuli, which have achieved dramatic results in several cancers. The future of cancer therapies is likely to combine these approaches with other treatments, including conventional therapies, to generate more effective treatments.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Regulação Neoplásica da Expressão Gênica/imunologia , Imunoterapia/métodos , Proteínas de Neoplasias/imunologia , Neoplasias/terapia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antineoplásicos/uso terapêutico , Terapia Combinada , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Transdução de Sinais , Resultado do Tratamento
8.
J Hepatol ; 62(1): 156-64, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25111176

RESUMO

BACKGROUND & AIMS: S100A4 has been linked to the fibrosis of several organs due to its role as a fibroblast-specific marker. However, the role of S100A4 itself in the development of fibrosis has not been much investigated. Here, we determined whether S100A4 regulates liver fibrogenesis and examined its mechanism by focusing on the activation of hepatic stellate cells (HSCs). METHODS: S100A4 deficient mice were used to determine the role of S100A4 in liver fibrogenesis. The effect of S100A4 on HSC activation was estimated by using primary mouse HSCs and the human HSC cell line LX-2. Serum levels of S100A4 in cirrhotic patients were determined by ELISA. RESULTS: S100A4 was found to be secreted by a subpopulation of macrophages and to promote the development of liver fibrosis. It accumulated in the liver during the progression of liver fibrosis and activated HSCs in mice. In vitro studies demonstrated that S100A4 induced the overexpression of alpha-smooth muscle actin through c-Myb in HSCs. Both, the selective depletion of S100A4-expressing cells and knockdown of S100A4 in the liver by RNA interference, resulted in a reduction of liver fibrosis following injury. Importantly, increased S100A4 levels in both the liver tissue and serum correlated positively with liver fibrosis in humans. CONCLUSIONS: S100A4 promotes liver fibrosis by activating HSCs, which may represent a potential target for anti-fibrotic therapies.


Assuntos
DNA/genética , Regulação da Expressão Gênica , Cirrose Hepática/genética , Proteínas S100/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Células Estreladas do Fígado/patologia , Humanos , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/biossíntese
9.
Cancer Immunol Immunother ; 64(8): 965-76, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25944005

RESUMO

BACKGROUND: Malignant pleural effusion (MPE) is a common complication caused by malignant diseases. However, subjectivity, poor sensitivity, and substantial false-negative rates of cytology assay hamper accurate MPE diagnosis. The aim of this study was to assess whether CD163+CD14+ tumor-associated macrophages (TAMs) could be used as a biomarker for enabling sensitive and specific MPE diagnosis. METHODS: Pleural effusion samples and peripheral blood samples were collected from 50 MPE patients and 50 non-malignant pleural effusion (NMPE) patients, respectively. Flow cytometry was performed to analyze cell phenotypes, and RT-qPCR was used to detect cytokine expression in these monocytes and macrophages. A blinded validation study (n = 40) was subsequently performed to confirm the significance of CD163+CD14+ TAMs in MPE diagnosis. Student's t test, rank sum test, and receiver operating characteristic curve analysis were used for statistical analysis. RESULTS: Notably, CD163+CD14+ cell frequency in MPE was remarkably higher than that in NMPE (P < 0.001). In a blinded validation study, a sensitivity of 78.9 % and a specificity of 100 % were obtained with CD163+CD14+ TAMs as a MPE biomarker. In total (n = 140), by using a cutoff level of 3.65 %, CD163+CD14+ cells had a sensitivity of 81.2 % and a specificity of 100 % for MPE diagnosis. Notably, MPE diagnosis by estimating CD163+CD14+ cells in pleural effusion could be obtained one week earlier than that obtained by cytological examination. CONCLUSIONS: CD163+CD14+ macrophages could be potentially used as an immune diagnostic marker for MPE and has better assay sensitivity than that of cytological analysis.


Assuntos
Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Biomarcadores Tumorais/análise , Receptores de Lipopolissacarídeos/análise , Macrófagos/imunologia , Monócitos/imunologia , Derrame Pleural Maligno/diagnóstico , Receptores de Superfície Celular/análise , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Biomarcadores Tumorais/imunologia , Separação Celular , Citocinas/metabolismo , Diagnóstico Diferencial , Reações Falso-Positivas , Feminino , Citometria de Fluxo , Humanos , Imunofenotipagem , Receptores de Lipopolissacarídeos/imunologia , Masculino , Pessoa de Meia-Idade , Derrame Pleural Maligno/imunologia , Receptores de Superfície Celular/imunologia , Sensibilidade e Especificidade
10.
J Hepatol ; 61(6): 1297-303, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25016226

RESUMO

BACKGROUND & AIMS: Liver cancer has a very dismal prognosis due to lack of effective therapy. Here, we studied the therapeutic effects of hyper-interleukin15 (hyper-IL-15), which is composed of IL-15 and the sushi domain of the IL-15 receptor α chain, on metastatic and autochthonous liver cancers. METHODS: Liver metastatic tumour models were established by intraportally injecting syngeneic mice with murine CT26 colon carcinoma cells or B16-OVA melanoma cells. Primary hepatocellular carcinoma (HCC) was induced by diethylnitrosamine (DEN). A hydrodynamics-based gene delivery method was used to achieve sustained hyper-IL-15 expression in the liver. RESULTS: Liver gene delivery of hyper-IL-15 robustly expanded CD8(+) T and NK cells, leading to a long-term (more than 40 days) accumulation of CD8(+) T cells in vivo, especially in the liver. Hyper-IL-15 treatment exerted remarkable therapeutic effects on well-established liver metastatic tumours and even on DEN-induced autochthonous HCC, and these effects were abolished by depletion of CD8(+) T cells but not NK cells. Hyper-IL-15 triggered IL-12 and interferon-γ production and reduced the expression of co-inhibitory molecules on dendritic cells in the liver. Adoptive transfer of T cell receptor (TCR) transgenic OT-1 cells showed that hyper-IL-15 preferentially expanded tumour-specific CD8(+) T cells and promoted their interferon-γ synthesis and cytotoxicity. CONCLUSIONS: Liver delivery of hyper-IL-15 provides an effective therapy against well-established metastatic and autochthonous liver cancers in mouse models by preferentially expanding tumour-specific CD8(+) T cells and promoting their anti-tumour effects.


Assuntos
Linfócitos T CD8-Positivos/patologia , Proliferação de Células/efeitos dos fármacos , Interleucina-15/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Metástase Neoplásica/tratamento farmacológico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Interferon gama/metabolismo , Interleucina-12/metabolismo , Interleucina-15/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/patologia , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica/patologia , Proteínas Recombinantes de Fusão/farmacologia , Resultado do Tratamento
11.
Life Sci ; 336: 122254, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37977355

RESUMO

AIMS: Gliomas are the most common central nervous system malignancies, with limited therapeutic options and poor prognosis, which are primarily attributed to the "immune desert" microenvironment. Previously, we constructed a three-gene-deleted oncolytic adenovirus (Ad-TD) loaded with non-secreting interleukin-12 (nsIL-12), which could be amplified in tumor cells and induce immunity to suppress tumors. However, the effects of this oncolytic virus on gliomas and their immune microenvironment remain unclear. There is an urgent need for further research. MATERIALS AND METHODS: We constructed a Syrian hamster brain tumor model and demonstrated the efficacy and mechanism of the novel oncolytic virus in treating brain tumors through a series of in vitro and in vivo experiments. We investigated the efficacy and safety (the number of hamsters in each group is either 5 or 10) of the oncolytic virus treatment in Syrian hamsters using a virus-treated group, a control virus-treated group, and a blank control group. KEY FINDINGS: In vitro assays showed that Ad-TD-nsIL-12 could specifically proliferate in brain tumor cells which induce tumor cell apoptosis and intracellular expression of interleukin (IL)-12. Moreover, in vivo experiments demonstrated that Ad-TD-nsIL-12 could effectively inhibit the progression of brain tumors and prolong survival. Ad-TD-nsIL-12 significantly enhanced T-cell infiltration in the brain tumor microenvironment. SIGNIFICANCE: Ad-TD-nsIL-12 can inhibit glioma progression and increase T-cell infiltration in the tumor tissue, particularly infiltration by cytotoxic T cells (CD8+). Ad-TD-nsIL-12 can amplify and produce IL-12, inducing anti-glioma immune responses to inhibit tumor progression.


Assuntos
Neoplasias Encefálicas , Glioma , Terapia Viral Oncolítica , Vírus Oncolíticos , Cricetinae , Animais , Humanos , Vírus Oncolíticos/genética , Interleucina-12/genética , Microambiente Tumoral , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Glioma/terapia , Neoplasias Encefálicas/terapia , Mesocricetus
12.
J Immunol ; 187(4): 1537-41, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21768399

RESUMO

Upon interaction with B7 homolog 1, programmed death-1 (PD-1) transmits a critical coinhibitory signal to T cells to negatively regulate immune responses. By extensively searching the genomic database with the IgV region of PD-1, we identified a homolog and named it PD-1 homolog (PD-1H). PD-1H is broadly expressed on the cell surface of hematopoietic cells and could be further upregulated on CD4(+) and CD8(+) T cells following activation. We have generated an mAb against PD-1H, which strikingly prevents acute graft-versus-host disease in semi- and fully allogeneic murine models, leading to full chimerism following treatment. Graft-versus-host disease remains a primary hindrance to successful allogeneic hematopoietic cell transplantation therapy for the treatment of hematologic malignancy. Therefore, manipulation of PD-1H function may provide a new modality for controlling T cell responses to allogeneic tissues in transplant medicine.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Antígenos de Superfície/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas , Animais , Antígenos de Superfície/genética , Proteínas Reguladoras de Apoptose/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/terapia , Humanos , Camundongos , Receptor de Morte Celular Programada 1 , Transplante Homólogo
13.
Front Immunol ; 14: 1097880, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36969175

RESUMO

Microglia/macrophages make up the largest population of tumor-infiltrating cells. Numerous studies have demonstrated that glioma-associated microglia/macrophages (GAMs) could promote the malignant progression of gliomas in various pathways. However, the primary function of GAMs in glioma remains inconclusive. First, by the CIBERSORT algorithm, we evaluated the content of microglia/macrophages in glioma tissues by bioinformatic analysis of omic data from thousands of glioma samples. Subsequently, we analyzed and confirmed the significant relationship between GAMs and the malignant phenotype of glioma, including survival time, IDH mutation status, and time of symptom onset. Afterward, Epithelial-Mesenchymal Transition (EMT) was identified by Gene Set Enrichment Analysis (GSEA) from numerous biological processes as the most relevant mechanism of malignant progression to GAMs. Moreover, a series of clinical samples were detected, including normal brain and various-grade glioma tissues. The results not only showed that GAMs were significantly associated with gliomas and their malignancy but also that GAMs were highly correlated with the degree of EMT in gliomas. In addition, we isolated GAMs from glioma samples and constructed co-culture models (in vitro) to demonstrate the promotion of the EMT process in glioma cells by GAMs. In conclusion, our study clarified that GAMs exert oncogenic effects with EMT in gliomas, suggesting the possibility of GAMs as immunotherapeutic targets.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Microglia/metabolismo , Neoplasias Encefálicas/metabolismo , Transição Epitelial-Mesenquimal , Microambiente Tumoral , Glioma/metabolismo , Macrófagos/metabolismo
14.
Curr Top Microbiol Immunol ; 344: 245-67, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20582531

RESUMO

Cancer immunotherapy is finally entering a new era with manipulation of cosignaling pathways as a therapeutic approach, for which the principle was proved nearly two decades ago. In addition to CTLA-4, CD137 and B7-H1/PD-1 pathways are two new targets in the stage. CD137 pathway is costimulatory and its agonistic antibody delivers potent signal to drive T cell growth and activation. On the other hand, blockade of B7-H1/PD-1 pathway with antagonistic antibody has shown to protect ongoing T cell responses from impairment by immune evasion mechanism in cancer microenvironment. With these tools in hand, a mechanism-based design of combined immunotherapy with high efficacy is becoming a reality.


Assuntos
Antígenos CD/fisiologia , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Neoplasias/terapia , Transdução de Sinais/fisiologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Transferência Adotiva , Animais , Proteínas Reguladoras de Apoptose/fisiologia , Antígeno B7-H1 , Humanos , Neoplasias/imunologia , Receptor de Morte Celular Programada 1 , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/fisiologia
15.
J Immunol ; 185(12): 7654-62, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21059892

RESUMO

Chronic hepatitis B virus (HBV) infection is characterized by sustained liver inflammation with an influx of lymphocytes, which contributes to the development of cirrhosis and hepatocellular carcinoma. The mechanisms underlying this immune-mediated hepatic pathogenesis remain ill defined. We report in this article that repetitive infusion of anti-CD137 agonist mAb in HBV-transgenic mice closely mimics this process by sequentially inducing hepatitis, fibrosis, cirrhosis, and, ultimately, liver cancer. CD137 mAb initially triggers hepatic inflammatory infiltration due to activation of nonspecific CD8(+) T cells with memory phenotype. CD8(+) T cell-derived IFN-γ plays a central role in the progression of chronic liver diseases by actively recruiting hepatic macrophages to produce fibrosis-promoting cytokines and chemokines, including TNF-α, IL-6, and MCP-1. Importantly, the natural ligand of CD137 was upregulated significantly in circulating CD14(+) monocytes in patients with chronic hepatitis B infection and closely correlated with development of liver cirrhosis. Thus, sustained CD137 stimulation may be a contributing factor for liver immunopathology in chronic HBV infection. Our studies reveal a common molecular pathway that is used to defend against viral infection but also causes chronic hepatic diseases.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Carcinoma Hepatocelular/imunologia , Vírus da Hepatite B/imunologia , Neoplasias Hepáticas Experimentais/imunologia , Monócitos/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Vírus da Hepatite B/genética , Hepatite B Crônica/genética , Hepatite B Crônica/imunologia , Hepatite B Crônica/metabolismo , Humanos , Memória Imunológica/efeitos dos fármacos , Memória Imunológica/genética , Memória Imunológica/imunologia , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Hepáticas Experimentais/virologia , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Monócitos/patologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/agonistas , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/biossíntese , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Regulação para Cima/imunologia
16.
Exp Mol Med ; 54(6): 765-776, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35672449

RESUMO

Classical monocytes are commonly involved in the innate inflammatory response and are the progenitors of osteoclasts. Excess endogenous glucocorticoids (GCs) can increase the levels of classical monocytes in blood and bone marrow. The role of this cell population in high-dose exogenous GC-induced osteoporosis (GIOP) remains to be elucidated. In this study, GIOP was established in rats and mice by daily methylprednisolone injection, and monocyte subsets were analyzed by flow cytometry. We demonstrated that classical monocytes accumulate in bone marrow during GIOP. Similarly, the monocyte proportion among bone marrow nucleated cells was also increased in patients with steroid treatment history. We sorted classical monocytes and analyzed their transcriptional profile in response to GCs by RNA sequencing. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that classical monocytes isolated from GC-treated rats exhibited osteoclast differentiation potential. Deletion of classical monocytes by clodronate liposome treatment prevented GIOP via inhibition of osteoclastogenesis and restoration of CD31HiendomucinHi vessels. Regarding the molecular mechanism, classical monocytes express high levels of glucocorticoid receptors. In vitro treatment with GCs increased both the percentage and absolute number of monocytes and promoted their proliferation. In summary, classical monocytes mediated GC-induced bone loss and are a potential target for therapeutic intervention in GIOP treatment.


Assuntos
Glucocorticoides , Osteoporose , Animais , Glucocorticoides/efeitos adversos , Camundongos , Monócitos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Osteoporose/induzido quimicamente , Ratos
17.
Cancer Immunol Res ; 10(7): 844-855, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35580259

RESUMO

High-mobility group protein B1 (HMGB1) is a danger signaling molecule that has been found to trigger an effective antitumor immune response. However, the mechanisms underlying its antitumor effects are not fully understood. Here, we found that HMGB1 release induced by chemotherapy in patients with non-small cell lung cancer was negatively correlated with PD-1 expression on CD8+ T cells. In vitro analysis indicated that treatment with HMGB1 led to a significant decrease in the level of expression of PD-1 on CD8+ T cells. Further analysis demonstrated that HMGB1 reduced PD-1 expression by inducing dynamin-mediated internalization of the protein, leading to early endocytosis in the cytoplasm, and subsequently degradation in the lysosomes. In a xenograft model, HER2-targeted chimeric antigen receptor (CAR) T cells had enhanced function in the presence of HMGB1. These data identify a role for HMGB1 as a negative regulator of PD-1 signaling in lung cancer and the observed antitumor effect of HMGB1 on CAR T cells may provide a theoretical foundation for a new immunotherapy combination.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Proteína HMGB1/metabolismo , Neoplasias Pulmonares , Linfócitos T CD8-Positivos , Carcinoma Pulmonar de Células não Pequenas/terapia , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/terapia , Receptor de Morte Celular Programada 1/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Blood ; 113(23): 5811-8, 2009 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-19339692

RESUMO

Programmed death one (PD-1) is an inducible molecule belonging to the immunoglobulin superfamily. It is expressed on activated T and B lymphocytes and plays pivotal roles in the negative regulation of adaptive immune responses. We report here an unexpected finding: that PD-1 could also be induced on splenic dendritic cells (DCs) by various inflammatory stimuli. Adoptive transfer of PD-1-deficient DCs demonstrates their superior capacity to wild-type DCs in innate protection of mice against lethal infection by Listeria monocytogenes. Furthermore, PD-1-deficient mice are also more resistant to the infection than wild-type controls, even in the absence of T and B cells, accompanied by elevated production of DC-derived interleukin-12 and tumor necrosis factor-alpha. Our results reveal a novel role of PD-1 in the negative regulation of DC function during innate immune response.


Assuntos
Antígenos de Diferenciação/imunologia , Células Dendríticas/imunologia , Imunidade Inata/imunologia , Listeriose/imunologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Linhagem Celular , Interleucina-12/imunologia , Ligantes , Listeria monocytogenes/imunologia , Camundongos , Camundongos Knockout , Receptor de Morte Celular Programada 1 , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Regulação para Cima/imunologia
19.
Int J Lab Hematol ; 43(2): 329-335, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33099889

RESUMO

INTRODUCTION: Monitoring of laboratory indicators is important for predicting changes in disease severity and clinical outcomes. We aimed to identify the critical predictors that can effectively assess the disease conditions of patients with COVID-19 by analyzing the clinical characteristics and laboratory findings of patients with SARS-CoV-2 infection. METHODS: All consecutive patients (n = 294) with confirmed SARS-CoV-2 infection admitted to the General Hospital of Central Theater Command of the PLA from February 6 to February 21, 2020, were enrolled. These patients were divided into the severe group and the nonsevere group according to disease severity during hospitalization. RESULTS: The median neutrophil-to-lymphocyte ratio (NLR) value of the severe patients was dramatically higher than that of the nonsevere patients (10.4 vs 2.6; P < .001). The NLR value equal to 5 was a boundary value worthy of reference, because more than 80% severe patients had an NLR value greater than 5 and over 80% nonsevere patients had an NLR value less than 5. The NLR value of these COVID-19 patients was positively and respectively correlated with the values of C-reactive protein (R = .5921, P < .001), lactate dehydrogenase (R = .4509, P < .001), procalcitonin (R = .5504, P < .001), fibrinogen (R = .4710, P < .001), and D-dimers (R = .4425, P < .001). However, the NLR value was merely and positively correlated with the interleukin-6 value (R = .3594, P < .05), but had no correlations with the values of interleukin-10, interleukin-4, interleukin-17, interferon-γ, and tumor necrosis factor-α (P > .05). DISCUSSION: Neutrophil-to-lymphocyte ratio is a critical predictor for assessment of disease severity in patients with COVID-19, and it has a close relation with the laboratory indicators related to disease conditions.


Assuntos
Proteína C-Reativa/metabolismo , COVID-19/diagnóstico , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Neutrófilos/patologia , SARS-CoV-2/patogenicidade , Linfócitos T/patologia , Adolescente , Adulto , Fatores Etários , Idoso , Biomarcadores/sangue , COVID-19/sangue , COVID-19/patologia , COVID-19/virologia , Feminino , Fibrinogênio/metabolismo , Humanos , Interleucina-6/sangue , L-Lactato Desidrogenase/sangue , Masculino , Pessoa de Meia-Idade , Neutrófilos/imunologia , Neutrófilos/virologia , Valor Preditivo dos Testes , Pró-Calcitonina/sangue , Estudos Retrospectivos , SARS-CoV-2/imunologia , Índice de Gravidade de Doença , Fatores Sexuais , Linfócitos T/imunologia , Linfócitos T/virologia
20.
Nat Commun ; 12(1): 951, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33574265

RESUMO

T cell rejuvenation by PD-1/PD-L1 blockade, despite emerging as a highly promising therapy for advanced cancers, is only beneficial for a minority of treated patients. There is evidence that a lack of efficient T cell activation may be responsible for the failure. Here, we demonstrate that IL-21 can be targeted to tumor-reactive T cells by fusion of IL-21 to anti-PD-1 antibody. To our surprise, the fusion protein PD-1Ab21 promotes the generation of memory stem T cells (TSCM) with enhanced cell proliferation. PD-1Ab21 treatment show potent antitumor effects in established tumor-bearing mice accompanied with an increased frequency of TSCM and robust expansion of tumor-specific CD8+ T cells with a memory phenotype, and is superior to a combination of PD-1 blockade and IL-21 infusion. Therefore, we have developed a potential strategy to improve the therapeutic effects of immune checkpoint blockade by simultaneously targeting cytokines to tumor-reactive T cells.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Memória Imunológica/efeitos dos fármacos , Interleucinas/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Linfócitos T , Animais , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Humanos , Ativação Linfocitária , Linfócitos do Interstício Tumoral , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/genética , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa