Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Oncoimmunology ; 13(1): 2293511, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38125721

RESUMEN

Anti-PD-1 antibody therapy has achieved success in tumor treatment; however, the duration of its clinical benefits are typically short. The functional state of intratumoral CD8+ T cells substantially affects the efficacy of anti-PD-1 antibody therapy. Understanding how intratumoral CD8+ T cells change will contribute to the improvement in anti-PD-1 antibody therapy. In this study, we found that tumor growth was not arrested after the late administration of anti-PD-1 antibody and that the antitumor function of CD8+ T cells decreased with tumor progression. The results of the RNA sequencing of CD8+ T cells infiltrating the tumor site on days 7 and 14 showed that the cell adhesion molecule Lymphocyte Function-associated Antigen-1 (LFA-1) participates in regulating the antitumor function of CD8+ T cells and that decreased LFA-1 expression in intratumoral CD8+ T cells is associated with tumor progression. By analyzing the Gene Expression Omnibus (GEO) database and our results, we found that the antitumor function of intratumoral CD8+ T cells with high LFA-1 expression was stronger. The formation of immune synapses is impaired in Itgal-si CD8+ T cells, resulting in decreased anti-tumor function. LFA-1 expression in intratumoral CD8+ T cells is regulated by the IL-2/STAT5 pathway. The combination of IL-2 and anti-PD-1 antibody effectively enhanced LFA-1 expression and the antitumor function of intratumoral CD8+ T cells. The adoptive transfer of OT-1 T cells overexpressing LFA-1, STAT5A, or STAT5B resulted in higher antitumor function, deferred tumor growth, and prolonged survival. These findings indicate that LFA-1-mediated immune synapse acts as a regulator of the antitumor function of intratumoral CD8+ T cells, which can be applied to improve anti-PD-1 antibody therapy.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Interleucina-2/farmacología , Factor de Transcripción STAT5/metabolismo , Moléculas de Adhesión Celular
2.
Cytotherapy ; 25(2): 202-209, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36379882

RESUMEN

BACKGROUND AIMS: Non-small cell lung cancer (NSCLC) remains the most common cancer worldwide, with an annual incidence of around 1.3 million. Surgery represents the standard treatment in early-stage NSCLC when feasible. However, because of cancer recurrence, only approximately 53% of patients with stage I and II NSCLC survive 5 years after radical surgery. The authors performed a retrospective study to investigate the impact of cytokine-induced killer (CIK) cell immunotherapy on the long-term survival of patients with stage I-II NSCLC after curative resection. METHODS: Fifty-seven patients with NSCLC were included in the study, with 41 and 16 in the control and CIK groups, respectively. Clinical characteristics were compared using a t-test and χ2 test. Survival analysis of patients with NSCLC was performed using the Kaplan-Meier method. The phenotypes and anti-tumor functions of CIK cells were evaluated by flow cytometry. RESULTS: Patients in the CIK group exhibited significantly longer overall survival (OS) and better disease-free survival (DFS) than those in the control group. Subgroup analysis indicated that patients with a higher risk of recurrence benefited more from CIK treatment and attained longer OS and DFS compared with those in the control group. No severe adverse events related to CIK treatment occurred. CIK cells contained a higher proportion of CD3+CD56+ natural killer (NK) T cells and CD3+ and CD8+ T cells and a lower proportion of CD3-CD56+ NK cells compared with peripheral blood mononuclear cells. CIK cells exhibited potent tumor-killing ability, with longer contact times with tumor cells and a greater number of cells exposed to tumor cells. CONCLUSIONS: The authors' data suggest that adjuvant CIK cell therapy is a safe and effective therapeutic strategy for improving OS and DFS in patients with stage I-II NSCLC after curative resection.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Células Asesinas Inducidas por Citocinas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Estudios Retrospectivos , Linfocitos T CD8-positivos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Recurrencia Local de Neoplasia/etiología , Inmunoterapia/efectos adversos
3.
Front Immunol ; 13: 1016646, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36238278

RESUMEN

Colorectal cancer (CRC) is the third most common cancer in the world. Although there are standard treatment options for CRC, most patients respond poorly to these treatments. Immunotherapies have gradually emerged due to the increasing awareness and understanding of tumor immunity, exhibiting good therapeutic efficacy in various cancers. Immunotherapies include cytokines, immune checkpoint inhibitors (ICIs), and adoptive cell therapies. In particular, ICIs, which are antibodies against cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), or its ligand PD-L1, have been successfully applied clinically for solid tumors, relieving the inhibitory effect of the tumor microenvironment on T cells. However, only a minority of patients with cancer achieve a durable clinical response during immunotherapy. Several factors restrict the efficacy of immunotherapy, leading to the development of drug resistance. In this review, we aimed to discuss the current status of immunotherapy for CRC and elaborate on the mechanisms that mediate resistance to immunotherapy and other potential therapeutic strategies.


Asunto(s)
Antígeno B7-H1 , Neoplasias Colorrectales , Antígeno CTLA-4 , Neoplasias Colorrectales/terapia , Citocinas , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Factores Inmunológicos , Inmunoterapia , Ligandos , Receptor de Muerte Celular Programada 1 , Microambiente Tumoral
4.
Cells ; 11(19)2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36231064

RESUMEN

T cells play central roles in the anti-tumor immunity, whose activation and differentiation are profoundly regulated by intrinsic metabolic reprogramming. Emerging evidence has revealed that metabolic processes of T cells are generally altered by tumor cells or tumor released factors, leading to crippled anti-tumor immunity. Therefore, better understanding of T cell metabolic mechanism is crucial in developing the next generation of T cell-based anti-tumor immunotherapeutics. In this review, we discuss how metabolic pathways affect T cells to exert their anti-tumor effects and how to remodel the metabolic programs to improve T cell-mediated anti-tumor immune responses. We emphasize that glycolysis, carboxylic acid cycle, fatty acid oxidation, cholesterol metabolism, amino acid metabolism, and nucleotide metabolism work together to tune tumor-reactive T-cell activation and proliferation.


Asunto(s)
Neoplasias , Linfocitos T , Aminoácidos/metabolismo , Ácidos Carboxílicos/metabolismo , Colesterol/metabolismo , Ácidos Grasos/metabolismo , Humanos , Neoplasias/metabolismo , Nucleótidos/metabolismo
5.
Cytotherapy ; 24(5): 526-533, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35219583

RESUMEN

BACKGROUND AIMS: In this retrospective clinical study, the authors investigated the impact of cytokine-induced killer (CIK) cell-based immunotherapies on the long-term survival of patients with esophageal squamous cell carcinoma (ESCC). METHODS: A total of 87 patients with ESCC who received comprehensive treatment were enrolled in the study. Of these patients, 43 were in the control group and 44 were in the CIK treatment group. Flow cytometry analysis was performed to detect the phenotype and anti-tumor function of CIK cells. Clinical characteristics were compared between these two groups, and the survival estimates of ESCC patients were determined using Kaplan-Meier analysis. RESULTS: CIK cells contained a high proportion of the main functional fraction (CD3+CD56+ group) and exhibited a strong killing ability for esophageal cancer cells in vitro. Importantly, overall survival (OS) and progression-free survival (PFS) were significantly higher in the CIK group than in the control group in early-stage ESCC. However, patients with advanced-stage ESCC did not benefit from CIK cell-based therapy in terms of OS and PFS compared with the control group. CONCLUSIONS: These results demonstrate that CIK cells combined with conventional treatments potentially prolong long-term survival of patients and may serve as a combined therapeutic approach for the treatment of early-stage ESCC.


Asunto(s)
Células Asesinas Inducidas por Citocinas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Terapia Combinada , Neoplasias Esofágicas/terapia , Carcinoma de Células Escamosas de Esófago/terapia , Humanos , Inmunoterapia , Inmunoterapia Adoptiva/métodos , Estudios Retrospectivos , Resultado del Tratamiento
7.
BMC Med ; 19(1): 283, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34819055

RESUMEN

BACKGROUND: Chimeric antigen receptor T (CAR-T) cell therapy has limited effects in the treatment of solid tumors. Sulforaphane (SFN) is known to play an important role in inhibiting tumor growth, but its effect on CAR-T cells remains unclear. The goal of the current study was to determine whether combined CAR-T cells and SFN could provide antitumor efficacy against solid tumors. METHODS: The effect of combined SFN and CAR-T cells was determined in vitro using a co-culture system and in vivo using a xenograft mouse model. We further validated the effects of combination therapy in patients with cancer. RESULTS: In vitro, the combination of SFN and CAR-T cells resulted in enhanced cytotoxicity and increased lysis of tumor cells. We found that SFN suppressed programmed cell death 1 (PD-1) expression in CAR-T cells and potentiated antitumor functions in vitro and in vivo. As a ligand of PD-1, programmed cell death ligand 1 (PD-L1) expression was also decreased in tumor cells after SFN treatment. In addition, ß-TrCP was increased by SFN, resulting in higher activation of ubiquitination-mediated proteolysis of PD-L1, which induced PD-L1 degradation. The combination of SFN and CAR-T cell therapy acted synergistically to promote better immune responses in vivo compared with monotherapy. In clinical treatments, PD-1 expression was lower, and proinflammatory cytokine levels were higher in patients with various cancers who received CAR-T cells and took SFN orally than that in the control group. CONCLUSION: SFN improves the cytotoxicity of CAR-T cells by modulating the PD-1/PD-L1 pathway, which may provide a promising strategy for the combination of SFN with CAR-T cells for cancer immunotherapy.


Asunto(s)
Antígeno B7-H1 , Receptores Quiméricos de Antígenos , Animales , Línea Celular Tumoral , Humanos , Inmunidad , Isotiocianatos , Ratones , Receptor de Muerte Celular Programada 1 , Sulfóxidos , Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cell Death Dis ; 12(10): 856, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34545063

RESUMEN

The refractory of castration-resistant prostate cancer (CRPC) is mainly reflected in drug resistance. The current research on the resistance mechanism of CRPC is still in its infancy. In this study, we revealed for the first time the key role of LncRNA PCBP1-AS1 in CRPC drug resistance. Through detailed in vivo and in vitro studies, we found that PCBP1-AS1 may enhance the deubiquitination of AR/AR-V7 by stabilizing the USP22-AR/AR-V7 complex, thereby preventing AR/AR-V7 from being degraded through the ubiquitin-proteasome pathway. Targeting PCBP1-AS1 can significantly restore the drug sensitivity of enzalutamide-resistant tumors in vivo and in vitro. Our research further expands the function of LncRNA in castration-resistant prostate cancer, which may provide new potential for clinical diagnosis and targeted therapy.


Asunto(s)
Benzamidas/uso terapéutico , Resistencia a Antineoplásicos , Nitrilos/uso terapéutico , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , ARN Largo no Codificante/metabolismo , Receptores Androgénicos/metabolismo , Ubiquitinación , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones Desnudos , Modelos Biológicos , Nitrilos/farmacología , Fenotipo , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Dominios Proteicos , Estabilidad Proteica , Proteolisis , ARN Largo no Codificante/genética , Receptores Androgénicos/química , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación/efectos de los fármacos
9.
Front Immunol ; 12: 680055, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34194433

RESUMEN

Background: There is increasing evidence that group 2 innate lymphoid cells (ILC2s) play an essential role in allergy and parasitic infection. However, the role of ILC2s in human lung cancer remains unclear. Methods: ILC2s from peripheral blood mononuclear cells (PBMCs) obtained from healthy donors (HDs) and non-small cell lung cancer (NSCLC) patients, and NSCLC tumor tissues were analyzed via multicolor flow cytometry. ILC2s or CD14+ cells were sorted by fluorescence-activated cell sorting. qPCR and flow cytometry were performed to assess the gene and protein expression of the indicated molecules. M1-like and M2-like macrophages were induced from CD14+ monocytes in vitro. Results: ILC2s were significantly more enriched in PBMCs and tumor tissues from NSCLC patients than in HDs. After screening for the main immune checkpoint molecules, we found that PD-1 was upregulated in ILC2s in NSCLC patients. Functionally, PD-1high ILC2s from tumor tissues expressed higher levels of IL-4 and IL-13 regarding both mRNA and protein levels than PD-1low ILC2s. Furthermore, PD-1high ILC2s robustly boosted M2-like macrophage polarization in vitro, by secreting IL-4 and IL-13, while neutralization of IL-4 and IL-13 by antibodies abrogated M2-like macrophage polarization. Conclusion: ILC2s are enriched in NSCLC patients and upregulate PD-1 expression. Upregulation of PD-1 facilitates the immunosuppressive function of ILC2s. PD-1high ILC2s enhance M2-like macrophage polarization by secreting IL-4 and IL-13. PD-1 acts as a positive regulator of the immunosuppressive function of ILC2s in human NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/etiología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Inmunidad Innata , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Biomarcadores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Citocinas/metabolismo , Femenino , Humanos , Inmunomodulación , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Transducción de Señal
10.
Cancer Lett ; 518: 35-48, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34139285

RESUMEN

Drug resistance remains the major obstacle limiting the effectiveness of chemotherapy for esophageal squamous cell carcinoma (ESCC)[1]. However, how stromal cells cooperate with immune cells to contribute to drug resistance is not yet fully understood. In this study, we observed that monocytic myeloid-derived suppressor cells (M-MDSCs) were correlated with cisplatin resistance in patients with ESCC. Furthermore, CAFs promoted differentiation of monocytes into M-MDSCs phenotypically and functionally in vitro. Mechanically, both interleukin (IL)-6 and exosome-packed microRNA-21 (miR-21) secreted by CAFs synergistically promoted the generation of M-MDSCs via activating the signal transducing activator of transcription 3 (STAT3) by IL-6 in an autocrine manner. Combined blocking of IL-6 receptor and inhibition of miR-21 significantly reversed CAF-mediated M-MDSC generation. Notably, the effects of CAFs on M-MDSC induction were abolished by inhibiting STAT3 signaling. Functionally, CAF-induced M-MDSCs promoted drug resistance of tumor cells upon cisplatin treatment. Clinically, ESCC patients with high infiltration of CAFs and CD11b+ myeloid cells had unfavorable predicted overall survival both in our cohort and in TCGA data. Taken together, our study reveals a paracrine and autocrine of IL-6 caused by CAFs co-activate STAT3 signaling, promoting the generation of M-MDSCs, and highlights the important role of CAFs in cooperation with M-MDSCs in promoting drug resistance, thus providing potential opportunities for reversing drug resistance through inhibition of STAT3 signaling.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Resistencia a Antineoplásicos/fisiología , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/metabolismo , Monocitos/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Transducción de Señal/fisiología , Fibroblastos Asociados al Cáncer/patología , Diferenciación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Cisplatino/farmacología , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/patología , Exosomas/metabolismo , Exosomas/patología , Humanos , Interleucina-6/metabolismo , MicroARNs/metabolismo , Monocitos/patología , Células Mieloides/metabolismo , Células Mieloides/patología , Células Supresoras de Origen Mieloide/patología , Factor de Transcripción STAT3/metabolismo
11.
Biomed Res Int ; 2020: 8765028, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33150182

RESUMEN

Immunotherapy, especially based on chimeric antigen receptor (CAR) T cells, has achieved prominent success in the treatment of hematological malignancies. However, approximately 30-50% of patients will have disease relapse following remission after receiving CD19-targeting CAR-T cells, with failure of maintaining a long-term effect. Mechanisms underlying CAR-T therapy inefficiency consist of loss or modulation of target antigen and CAR-T cell poor persistence which mostly results from T cell exhaustion. The unique features and restoration strategies of exhausted T cells (Tex) have been well described in solid tumors. However, the overview associated with CAR-T cell exhaustion is relatively rare in hematological malignancies. In this review, we summarize the characteristics, cellular, and molecular mechanisms of Tex cells as well as approaches to reverse CAR-T cell exhaustion in hematological malignancies, providing novel strategies for immunotherapies.


Asunto(s)
Antígenos CD19/genética , Anergia Clonal , Neoplasias Hematológicas/terapia , Inmunoterapia Adoptiva/métodos , Receptores Quiméricos de Antígenos/genética , Linfocitos T/inmunología , Antígenos CD19/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Antígenos CD28/genética , Antígenos CD28/inmunología , Expresión Génica , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Humanos , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Recurrencia , Linfocitos T/patología , Insuficiencia del Tratamiento , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
12.
J Immunol ; 204(9): 2575-2588, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32221038

RESUMEN

Metformin has been studied for its anticancer effects by regulating T cell functions. However, the mechanisms through which metformin stimulates the differentiation of memory T cells remain unclear. We found that the frequencies of memory stem and central memory T cells increased for both in peripheral and tumor-infiltrating CD8+ T cells in metformin-treated lung cancer patients compared with those not taking the medication. An in vitro assay showed that metformin promoted the formation of memory CD8+ T cells and enhanced their antiapoptotic abilities. In addition, AMP-activated protein kinase (AMPK) activation decreased microRNA-107 expression, thus enhancing Eomesodermin expression, which suppressed the transcription of PDCD1 in metformin-treated CD8+ T cells. In the CAR-T cell therapy model, metformin also exhibited cytotoxicity-promoting effects that led to decreased tumor growth. Metformin could reprogram the differentiation of CD8+ T cells, which may benefit the clinical therapy of cancer patients by facilitating long-lasting cytotoxic functions.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Metformina/farmacología , MicroARNs/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas de Dominio T Box/metabolismo , Células A549 , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células HCT116 , Humanos , Memoria Inmunológica/efectos de los fármacos , Estudios Retrospectivos , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
13.
Cancer Manag Res ; 11: 7813-7824, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31695486

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors worldwide and the 5-year overall survival rate remains poor. Protein kinase, membrane associated tyrosine/threonine (PKMYT1) is overexpressed in several cancers and participate in tumor progression. However, the mechanism of PKMYT1 in ESCC is unclear. PURPOSE: The objective of our study was to demonstrate the the expression and role of PKMYT1 in ESCC. PATIENTS AND METHODS:   We detected the expression of PKMYT1 in ESCC patients and analysed the correlation with overall survival time and disease-free survival time. Then we detected PKMYT1 expression in ESCC cell lines and immortalized human esophageal epithelial cell line. Down-regulated PKMYT1 was carried out in KYSE70 and KYSE450 cells to invetigate the mechanism of PKMYT1 in ESCC cells. RESULTS: PKMYT1 was up-regulated in tumor tissues and ESCC cell lines, and higher expression of PKMYT1 correlated with poorer overall survival in ESCC patients. Besides, in ESCC cell lines KYSE70 and KYSE450, knocking down PKMYT1 allowed more cells to skip G2/M checkpoint to complete mitosis, which promoted cell apoptosis, inhibited cell proliferation, and prevented the EMT phenotype in vitro. Meantime, we also observed that down-regulated PKMYT1 in ESCC cells suppressed AKT/mTOR signaling pathway. These results demonstrated PKMYT1 may act as an oncogene in ESCC. CONCLUSION: PKMYT1 plays an crutial role in ESCC progression, downregulated PKMYT1 might inhibit the development of ESCC by AKT/mTOR signaling pathway, and might be a novel target in the treatment of ESCC.

14.
Cancer Manag Res ; 10: 6079-6096, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538557

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is a major cause of cancer mortality and an increasing incidence worldwide; however, there are very few effective diagnostic approaches and prognostic biomarkers. MATERIALS AND METHODS: One hundred forty-nine pairs of HCC samples from Gene Expression Omnibus (GEO) were obtained to screen differentially expressed genes (DEGs) between HCC and normal samples. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, Gene ontology enrichment analyses, and protein-protein interaction network were used. Cox proportional hazards regression analysis was used to identify significant prognostic DEGs, with which a gene expression signature prognostic prediction model was identified in The Cancer Genome Atlas (TCGA) project discovery cohort. The robustness of this panel was assessed in the GSE14520 cohort. We verified details of the gene expression level of the key molecules through TCGA, GEO, and qPCR and used immunohistochemistry for substantiation in HCC tissues. The methylation states of these genes were also explored. RESULTS: Ninety-eight genes, consisting of 13 upregulated and 85 downregulated genes, were screened out in three datasets. KEGG and Gene ontology analysis for the DEGs revealed important biological features of each subtype. Protein-protein interaction network analysis was constructed, consisting of 64 nodes and 115 edges. A subset of four genes (SPINK1, TXNRD1, LCAT, and PZP) that formed a prognostic gene expression signature was established from TCGA and validated in GSE14520. Next, the expression details of the four genes were validated with TCGA, GEO, and clinical samples. The expression panels of the four genes were closely related to methylation states. CONCLUSION: This study identified a novel four-gene signature biomarker for predicting the prognosis of HCC. The biomarkers may also reveal molecular mechanisms underlying development of the disease and provide new insights into interventional strategies.

15.
Int Immunopharmacol ; 51: 57-65, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28802902

RESUMEN

Pseudomonas aeruginosa-mannose sensitive hemagglutinin (PA-MSHA) injection serves as immunological adjuvant in clinical treatment of cancer patients. In present study, we investigated whether PA-MSHA injection enhanced the anti-tumor efficacy of CIK cells. Twenty patients with malignancies were enrolled in this retrospective clinical trial. They were divided into two groups: 10 patients received PA-MSHA treated CIK cells transfusion combined with chemotherapy, and other patients accepted CIK cells and chemotherapy. The efficacy of PA-MSHA treated CIK cells was also observed in vitro and in vivo. With PA-MSHA treatment CIK cells exhibited enhanced proliferation but decreased expression of inhibitory cell surface markers such as Tim-3 and PD-1. Particularly in CIK cells, PA-MSHA promoted the extrusion of pro-inflammatory cytokines like IFN-γ. Of 10 patients with PA-MSHA treated CIK cells and chemotherapy, two patients reached partial remissions, 7 patients had stable disease and the other one had progressive disease. Some of these patients experienced fever after cell infusion. 8 patients with CIK cells showed stable disease and 2 patients had progressive disease. Moreover, the side effects were small in patients with CIK treatment. Our data indicated that PA-MSHA improves the functions of CIK cells and shed new light on developing more potent therapeutic approaches for malignancies.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Neoplasias del Colon/terapia , Células Asesinas Inducidas por Citocinas/inmunología , Proteínas Fimbrias/administración & dosificación , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/terapia , Neoplasias Ováricas/terapia , Pseudomonas aeruginosa/inmunología , Adulto , Anciano , Células Cultivadas , Neoplasias del Colon/inmunología , Terapia Combinada , Células Asesinas Inducidas por Citocinas/trasplante , Quimioterapia , Femenino , Humanos , Interferón gamma/metabolismo , Neoplasias Pulmonares/inmunología , Masculino , Persona de Mediana Edad , Neoplasias Ováricas/inmunología , Inducción de Remisión , Estudios Retrospectivos
16.
Adolescence ; 44(176): 827-50, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20432603

RESUMEN

The Felder-Soloman Index of Learning Styles (ILS) has been a popular instrument for measuring learning styles of college students for the past two decades. Even though several researchers have translated the ILS into Chinese for their own studies, a Chinese version has not been standardized and evaluated, nor has anyone reported on its reliability and validity. Based on data collected from 2,748 students at a large private university in Taiwan, this study investigates the reliability and validity of the Chinese version of the ILS. In addition, through factor analysis and structural equation modeling (SEM) analysis, problematic test items are identified for further modification. Results show that the reliability of each scale of the ILS has a pattern similar to that of previous studies. The study therefore investigates the identified problematic elements and discusses two key points: (1) the language and translation problems and (2) precision and design. In addition, results of the significant interaction effects of analysis of variance (ANOVA) for active/reflective and sensing/intuitive scales indicate the effect of college differences depends on the levels between genders. Moreover, in general, female students are significantly more intuitive and global and less visual than male students. Other detailed analysis of academic disciplines and gender onILS are also reported.


Asunto(s)
Aprendizaje , Pruebas Psicológicas , Estudiantes/psicología , Adolescente , Análisis Factorial , Femenino , Humanos , Masculino , Psicometría , Reproducibilidad de los Resultados , Factores Sexuales , Taiwán
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...