Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Sci Rep ; 14(1): 15682, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38977778

RESUMEN

This study constructed a comprehensive analysis of cell death modules in eliminating aberrant cells and remodeling tumor microenvironment (TME). Consensus analysis was performed in 490 lung adenocarcinoma (LUAD) patients based on 4 types of cell death prognostic genes. Intersection method divided these LUAD samples into 5 cell death risk (CDR) clusters, and COX regression analysis were used to construct the CDR signature (CDRSig) with risk scores. Significant differences of TME phenotypes, clinical factors, genome variations, radiosensitivity and immunotherapy sensitivity were observed in different CDR clusters. Patients with higher risk scores in the CDRSig tended to be immune-excluded or immune-desert, and those with lower risk scores were more sensitive to radiotherapy and immunotherapy. The results from mouse model showed that intense expression of the high-risk gene PFKP was associated with low CD8+ T cell infiltration upon radiotherapy and anti-PD-L1 treatment. Deficient assays in vitro confirmed that PFKP downregulation enhanced cGAS/STING pathway activation and radiosensitivity in LUAD cells. In conclusion, our studies originally performed a comprehensive cell death analysis, suggesting the importance of CDR patterns in reprogramming TME and providing novel clues for LUAD personalized therapies.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Medicina de Precisión , Microambiente Tumoral , Microambiente Tumoral/inmunología , Humanos , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/terapia , Adenocarcinoma del Pulmón/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/patología , Medicina de Precisión/métodos , Animales , Ratones , Muerte Celular , Regulación Neoplásica de la Expresión Génica , Inmunoterapia/métodos , Línea Celular Tumoral , Pronóstico , Femenino , Masculino
2.
Zhongguo Zhen Jiu ; 44(6): 631-6, 2024 Jun 12.
Artículo en Chino | MEDLINE | ID: mdl-38867623

RESUMEN

OBJECTIVE: To compare the clinical effect of intradermal needling and acupuncture in prevention and treatment of leukopenia after chemotherapy with spleen-kidney deficiency. METHODS: A total of 90 patients with malignant tumor who received chemotherapy were randomly divided into a intradermal needling group (30 cases, 1 case dropped out), an acupuncture group (30 cases, 2 cases dropped out, 1 case was eliminated) and a control group (30 cases). The control group received conventional symptomatic treatment after chemotherapy. On the basis of the treatment in the control group, the intradermal needling group received intradermal needling at Guanyuan (CV 4), Dazhui (GV 14) and bilateral Geshu (BL 17), Zusanli (ST 36),Shenshu (BL 23), the needles were retained for 48 h, once every other day. On the basis of the treatment in the control group, the acupuncture group received conventional acupuncture at the same acupoints as the intradermal needling group, once every other day. The treatment started from the first day of chemotherapy, for a total of 2 weeks in the three groups. The white blood cell count, neutrophil count, hemoglobin content, platelet count and Karnofsky performance status (KPS) score before treatment and on 3rd, 7th, 14th, and 21st days after treatment were compared among the three groups. The incidence and grading of leukopenia and the usage of leukocyte-boosting drug during chemotherapy cycle was recorded. RESULTS: On 7th day after treatment, the white blood cell count in the intradermal needling group and the acupuncture group was higher than that in the control group (P<0.01, P<0.05). On the 14th day after treatment, the hemoglobin content in the intradermal needling group and the acupuncture group was higher than that in the control group (P<0.01). On the 7th, 14th, and 21st days after treatment, the platelet count in the acupuncture group was higher than that in the control group (P<0.01), on the 14th and 21st days after treatment, the platelet count in the intradermal needling group was higher than that in the control group (P<0.01). There was no statistically significant difference among the three groups after treatment in terms of neutrophil count, KPS score, incidence and grading of leukopenia, and the usage of leukocyte-boosting drug (P>0.05). CONCLUSION: Both intradermal needling and acupuncture can effectively increase peripheral blood white blood cell count, hemoglobin content and platelet count during chemotherapy cycle, reduce the toxicity of chemotherapy drug to bone marrow hematopoietic function, and alleviate bone marrow suppression after chemotherapy. The two treatments are equally effective.


Asunto(s)
Terapia por Acupuntura , Leucopenia , Humanos , Leucopenia/etiología , Leucopenia/prevención & control , Leucopenia/terapia , Masculino , Femenino , Persona de Mediana Edad , Adulto , Anciano , Bazo/efectos de los fármacos , Bazo/fisiopatología , Adulto Joven , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Antineoplásicos/efectos adversos , Riñón/fisiopatología , Riñón/efectos de los fármacos , Puntos de Acupuntura
3.
Cancer Immunol Immunother ; 73(4): 71, 2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38430394

RESUMEN

BACKGROUND: Due to individual differences in tumors and immune systems, the response rate to immunotherapy is low in lung adenocarcinoma (LUAD) patients. Combinations with other therapeutic strategies improve the efficacy of immunotherapy in LUAD patients. Although radioimmunotherapy has been demonstrated to effectively suppress tumors, the underlying mechanisms still need to be investigated. METHODS: Total RNA from LUAD cells was sequenced before and after radiotherapy to identify differentially expressed radiation-associated genes. The similarity network fusion (SNF) algorithm was applied for molecular classification based on radiation-related genes, immune-related genes, methylation data, and somatic mutation data. The changes in gene expression, prognosis, immune cell infiltration, radiosensitivity, chemosensitivity, and sensitivity to immunotherapy were assessed for each subtype. RESULTS: We used the SNF algorithm and multi-omics data to divide TCGA-LUAD patients into three subtypes. Patients with the CS3 subtype had the best prognosis, while those with the CS1 and CS2 subtypes had poorer prognoses. Among the strains tested, CS2 exhibited the most elevated immune cell infiltration and expression of immune checkpoint genes, while CS1 exhibited the least. Patients in the CS2 subgroup were more likely to respond to PD-1 immunotherapy. The CS2 patients were most sensitive to docetaxel and cisplatin, while the CS1 patients were most sensitive to paclitaxel. Experimental validation of signature genes in the CS2 subtype showed that inhibiting the expression of RHCG and TRPA1 could enhance the sensitivity of lung cancer cells to radiation. CONCLUSIONS: In summary, this study identified a risk classifier based on multi-omics data that can guide treatment selection for LUAD patients.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Multiómica , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/terapia , Inmunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Análisis por Conglomerados , Pronóstico
4.
Genet Test Mol Biomarkers ; 27(12): 393-405, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38156905

RESUMEN

Background: There is increasing evidence that abnormal expression of microRNAs is involved in the occurrence and progression of tumors. In previous experiments, we found that the content of hsa-miR-1301-3p in tumor tissues of patients with nonsmall cell lung cancer (NSCLC) showed an obvious upward trend compared with that in normal tissues. We performed a detailed study on the impact and underlying mechanism of hsa-miR-1301-3p in NSCLC cells. Methods: The impact of hsa-miR-1301-3p on NSCLC cell proliferation, apoptosis, migration, and invasion was examined using colony formation, flow cytometry, modified Boyden chamber, and wound healing assays. Different doses of radiation were applied to NSCLC cells to investigate their sensitivity to radiotherapy. The potential target gene of hsa-miR-1301-3p was determined by dual-luciferase reporter assay and immunoblotting. Result: hsa-miR-1301-3p was upregulated in NSCLC tissues and cells. hsa-miR-1301-3p effectively promoted the rapid proliferation, migration, and invasion of NSCLC cells, while inhibiting apoptosis. It also induced radioresistance in NSCLC cells. hsa-miR-1301-3p targeted the homeodomain-only protein homeobox (HOPX) mRNA 3' untranslated region and inhibited its transcription in NSCLC cells. Exogenous HOPX overexpression antagonized the mechanism by which hsa-miR-1301-3p regulates NSCLC cell proliferation, metastasis, and apoptosis. Conclusions: hsa-miR-1301-3p plays an oncogenic role in the occurrence and development of NSCLC. By targeting HOPX, hsa-miR-1301-3p can not only promote the proliferation and metastasis of NSCLC cells, but also alleviate apoptosis and reduce radiosensitivity.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , Humanos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Genes Homeobox , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Tolerancia a Radiación/genética
6.
Acad Radiol ; 30(10): 2156-2168, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37003875

RESUMEN

RATIONALE AND OBJECTIVES: To develop a multimodal ultrasound radiomics nomogram for accurate classification of thyroid micronodules. MATERIALS AND METHODS: A retrospective study including 181 thyroid micronodules within 179 patients was conducted. Radiomics features were extracted from strain elastography (SE), shear wave elastography (SWE) and B-mode ultrasound (BMUS) images. Minimum redundancy maximum relevance and least absolute shrinkage and selection operator algorithms were used to select malignancy-related features. BMUS, SE, and SWE radiomics scores (Rad-scores) were then constructed. Multivariable logistic regression was conducted using radiomics signatures along with clinical data, and a nomogram was ultimately established. The calibration, discriminative, and clinical usefulness were considered to evaluate its performance. A clinical prediction model was also built using independent clinical risk factors for comparison. RESULTS: An aspect ratio ≥ 1, mean elasticity index, BMUS Rad-score, SE Rad-score, and SWE Rad-score were identified as the independent predictors for predicting malignancy of thyroid micronodules by multivariable logistic regression. The radiomics nomogram based on these characteristics showed favorable calibration and discriminative capabilities (AUCs: 0.903 and 0.881 for training and validation cohorts, respectively), all outperforming clinical prediction model (AUCs: 0.791 and 0.626, respectively). The decision curve analysis also confirmed clinical usefulness of the nomogram. The significant improvement of net reclassification index and integrated discriminatory improvement indicated that multimodal ultrasound radiomics signatures might work as new imaging markers for classifying thyroid micronodules. CONCLUSION: The nomogram combining multimodal ultrasound radiomics features and clinical factors has the potential to be used for accurate diagnosis of thyroid micronodules in the clinic.


Asunto(s)
Diagnóstico por Imagen de Elasticidad , Neoplasias , Humanos , Modelos Estadísticos , Estudios Retrospectivos , Glándula Tiroides/diagnóstico por imagen , Pronóstico , Nomogramas
7.
Immunology ; 168(2): 346-361, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36326481

RESUMEN

Tumour-associated macrophages (TAMs) are one of the primary sources of PD-L1 expression in the tumour microenvironment (TME). Ionizing radiation (IR) promotes PD-L1 expression in tumour cells. However, the effect of IR on macrophage PD-L1 expression and the underlying mechanisms remain unclear. ATM kinase, as the key kinase for initiating DNA damage repair (DDR) process, is associated with innate immune STING axis activation. Here, we explored the molecular mechanism implicated in macrophage PD-L1 expression regulated by IR as well as the role of ATM kinase in this process. IR-regulated PD-L1 expression in macrophages and associated signalling pathways were explored by in vitro studies using murine and human macrophage cell lines. A colorectal xenograft murine model was employed to demonstrate the impact of targeting ATM and PD-L1 expression in TAMs following IR on growth of tumour in vivo. IR up-regulated PD-L1 expression in macrophages, which was further augmented by ATM kinase inhibition. ATM inhibition increased IR-induced DNA damage, which activated STING/interferon regulatory factor 3 (IRF3) signalling pathway and up-regulated type I interferon (IFN-I) expression in macrophages. IFN-I bound to the IFN α receptor 1 on macrophages, activated the downstream JAK1 and STAT1/3 signalling and eventually led to PD-L1 up-expression. ATM inhibition augmented IR-induced PD-L1 expression in macrophages and CD8+ T cell infiltration, and promoted anti-tumour efficacy in vivo. These results suggested that ATM inhibition promoted IR-induced PD-L1 expression through the activation of innate immunity in TAMs, which provided a novel approach to enhance the anti-tumour efficacy of RT.


Asunto(s)
Ataxia Telangiectasia , Neoplasias , Humanos , Animales , Ratones , Interferones , Macrófagos Asociados a Tumores , Antígeno B7-H1/metabolismo , Transducción de Señal , Microambiente Tumoral
9.
Cancers (Basel) ; 14(16)2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-36010926

RESUMEN

Cancer severely threatens human health and has remained the leading cause of disease-related death for decades. With the rapid advancement of nanomedicine, nanoscale metal-organic frameworks are believed to be potentially applied in the treatment and biomedical imaging for various tumors. Zeolite imidazole framework (ZIF)-8 attracts increasing attention due to its high porosity, large specific surface area, and pH-responsiveness. The designs and modifications of ZIF-8 nanoparticles, as well as the strategy of drug loading, demand a multifaceted and comprehensive understanding of nanomaterial features and tumor characteristics. We searched for studies on ZIF-8-based nanoplatforms in tumor theranostics on Web of Science from 2015 to 2022, mainly focused on the research published in the past 3 years, summarized the progress of their applications in tumor imaging and treatment, and discussed the favorable aspects of ZIF-8 nanoparticles for tumor theranostics as well as the future opportunities and potential challenges. As a kind of metal-organic framework material full of potential, ZIF-8 can be expected to be combined with more therapeutic systems in the future and continue to contribute to all aspects of tumor therapy and diagnosis.

10.
Front Genet ; 13: 935601, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36035179

RESUMEN

Lung adenocarcinoma (LUAD) has high morbidity and mortality worldwide, and its prognosis remains unsatisfactory. Identification of epigenetic biomarkers associated with radiosensitivity is beneficial for precision medicine in LUAD patients. SETD2 is important in repairing DNA double-strand breaks and maintaining chromatin integrity. Our studies established a comprehensive analysis pipeline, which identified SETD2 as a radiosensitivity signature. Multi-omics analysis revealed enhanced chromatin accessibility and gene transcription by SETD2. In both LUAD bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq), we found that SETD2-associated positive transcription patterns were associated with DNA damage responses. SETD2 knockdown significantly upregulated tumor cell apoptosis, attenuated proliferation and migration of LUAD tumor cells, and enhanced radiosensitivity in vitro. Moreover, SETD2 was a favorably prognostic factor whose effects were antagonized by the m6A-related genes RBM15 and YTHDF3 in LUAD. In brief, SETD2 was a promising epigenetic biomarker in LUAD patients.

11.
Cancer Lett ; 545: 215852, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35926817

RESUMEN

Small cell lung cancer (SCLC) is a highly malignant tumor with extremely poor prognosis. The treatment strategy is very limited, and patient outcomes remain dismal with the 5-year survival rate being mere 3-6%. Thus, novel therapeutic strategies for SCLC patients are urgently needed. In this study, we found that the triple-therapy of poly (ADP-ribose) polymerase (PARP) inhibitor, radiotherapy (RT) and anti-PD-1 treatment significantly inhibited tumor growth and prolonged survival in the syngeneic SCLC models in immunocompetent C57BL/6 mice. Mechanistically, we demonstrated that the combination of PARP inhibitor niraparib and RT reshaped an inflamed tumor microenvironment, including activation of the cGAS/STING immune response pathway, induction of immunogenic cell death, and upregulation of PD-L1 on tumor cells. Furthermore, this triple-therapy substantially augmented CD8+ T cell infiltration and activation, and enhanced anti-tumor effects as revealed by increased median survival time and reduced tumor volume without additional myelosuppression or hepatic injury. Together, our studies demonstrated that PARP inhibitor combined with RT potentiated anti-tumor immunity and enhanced the efficacy of anti-PD-1 immunotherapy in preclinical study, which provided a promising therapeutic strategy for SCLC patients in clinic.


Asunto(s)
Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Animales , Antígeno B7-H1 , Línea Celular Tumoral , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Ratones , Ratones Endogámicos C57BL , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Receptor de Muerte Celular Programada 1 , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/radioterapia , Microambiente Tumoral
12.
Commun Biol ; 5(1): 663, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35790819

RESUMEN

The purpose of this study is to examine the association between G protein-coupled receptor 87 (GPR87) and lung adenocarcinoma (LUAD) metastasis and immune infiltration. The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets extract clinical data. According to the TCGA database, increased GPR87 expression predicts poor overall survival, progression-free interval, and disease-specific survival in LUAD patients. The meta-analysis also reveals a significant association between high GPR87 expression and poor overall survival. Moreover, functional experiments demonstrate that GPR87 silencing reduces LUAD cell invasion and migration. Immunoblotting shows that GPR87 knockdown decreased Vimentin and N-cadherin expression and increased E-cadherin expression in LUAD cells. GPR87 expression in LUAD is positively correlated with immune infiltration. In addition, GPR87 expression is associated with immune and chemotherapy resistance in LUAD patients. Our findings indicate that GPR87 promotes tumor progression and is correlated with immune infiltration, suggesting GPR87 as a possible biomarker for prognosis prediction in LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Adenocarcinoma , Neoplasias Pulmonares , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Procesos Neoplásicos , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo
13.
Cancer Med ; 10(24): 8944-8960, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34741449

RESUMEN

Cancer stem cells (CSCs) have been shown to accelerate tumor recurrence, radiotherapy, and chemotherapy resistance. Immunotherapy is a powerful anticancer treatment that can significantly prolong the overall survival of patients with lung adenocarcinoma (LUAD). However, little is known about the function of genes related to tumor stemness and immune infiltration in LUAD. After integrating the tumor stemness index based on mRNA expression (mRNAsi), immune score, mRNA expression, and clinical information from the TCGA database, we screened 380 tumor stemness and immune (TSI)-related genes and constructed a five TSI-specific-gene (CPS1, CCR2, NT5E, ANLN, and ABCC2) signature (TSISig) using a machine learning method. Survival analysis indicated that TSISig could stably predict the prognosis of patients with LUAD. Comparison of mRNAsi and immune score between high- and low-TSISig groups suggested that TSISig characterized tumor stemness and immune infiltration. In addition, enrichment of immune subpopulations showed that the low-TSISig group held more immune subpopulations. GSEA revealed that TSISig had a strong association with the cell cycle and human immune response. Further analysis revealed that TSISig not only had a good predictive ability for prognosis but could also serve as an excellent predictor of tumor recurrence and response to radiotherapy and immunotherapy in LUAD patients. TSISig might regulate the development of LUAD by coordinating tumor stemness and immune infiltration. Finally, a connectivity map (CMap) analysis demonstrated that the HDAC inhibitor could target TSISig.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/radioterapia , Inmunoterapia/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Células Madre Neoplásicas/metabolismo , Adenocarcinoma del Pulmón/mortalidad , Humanos , Neoplasias Pulmonares/mortalidad , Análisis de Supervivencia
14.
Int J Oncol ; 59(5)2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34558640

RESUMEN

Targeted therapy with epidermal growth factor receptor (EGFR)­tyrosine kinase inhibitors (TKIs) is a standard modality of the 1st­line treatments for patients with advanced EGFR­mutated non­small cell lung cancer (NSCLC), and substantially improves their prognosis. However, EGFR T790M mutation is the primary mechanism of 1st­ and 2nd­generation EGFR­TKI resistance. Osimertinib is a representative of the 3rd­generation EGFR­TKIs that target T790M mutation, and has satisfactory efficacy in the treatment of T790M­positive NSCLC with disease progression following use of 1st­ or 2nd­generation EGFR­TKIs. Other 3rd­generation EGFR­TKIs, such as abivertinib, rociletinib, nazartinib, olmutinib and alflutinib, are also at various stages of development. However, the occurrence of acquired resistance is inevitable, and the mechanisms of 3rd­generation EGFR­TKI resistance are complex and incompletely understood. Genomic studies in tissue and liquid biopsies of resistant patients reveal multiple candidate pathways. The present review summarizes the recent findings in mechanisms of resistance to 3rd­generation EGFR­TKIs in advanced NSCLC, and provides possible strategies to overcome this resistance. The mechanisms of acquired resistance mainly include an altered EGFR signaling pathway (EGFR tertiary mutations and amplification), activation of aberrant bypassing pathways (hepatocyte growth factor receptor amplification, human epidermal growth factor receptor 2 amplification and aberrant insulin­like growth factor 1 receptor activation), downstream pathway activation (RAS/RAF/MEK/ERK and PI3K/AKT/mTOR) and histological/phenotypic transformations (SCLC transformation and epithelial­mesenchymal transition). The combination of targeted therapies is a promising strategy to treat osimertinib­resistant patients, and multiple clinical studies on novel combined therapies are ongoing.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteína 11 Similar a Bcl2/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Resistencia a Antineoplásicos , Receptores ErbB/genética , Receptores ErbB/fisiología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
Cancer Med ; 10(15): 5256-5269, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34152094

RESUMEN

BACKGROUND: Human endogenous retrovirus-H long terminal repeat-associating protein 2 (HHLA2) is a member of B7 family, which is upregulated in multiple tumors. However, its exact functions in non-small cell lung cancer (NSCLC) have not been fully understood. This study aimed to investigate the biological roles of HHLA2 in human NSCLC and the relevant mechanisms. In addition, the effects of tumor cell-derived HHLA2 on tumor-associated macrophage (TAM) polarization were explored. METHODS: NSCLC cell growth, migration, and invasion were assessed by colony formation and modified Boyden chamber assays. Cell cycle and the CD163+ TAMs were examined by flow cytometry. A co-culture model of THP-1 macrophages and NSCLC cells was conducted to investigate the impacts of tumor cell-derived HHLA2 on THP-1 macrophage polarization. Moreover, a xenograft nude mouse model was established to explore the effects of HHLA2 on tumorigenesis in vivo. RESULTS: HHLA2 was upregulated in A549 and H1299 cells compared with the normal lung epithelial BEAS-2B cells. HHLA2 deficiency inhibited NSCLC cell proliferation, migration, invasion, and induced G0/G1 phase arrest partially via inhibiting EGFR/MAPK/ERK signaling pathway. Furthermore, HHLA2 knockdown inhibited M2 polarization of TAMs via downregulating IL-10. In addition, knockdown of HHLA2 inhibited tumor growth in vivo. CONCLUSION: HHLA2 downregulation inhibited NSCLC growth and TAM M2 polarization. HHLA2 may serve as a therapeutic target and promising prognostic biomarker in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Polaridad Celular , Progresión de la Enfermedad , Inmunoglobulinas/deficiencia , Neoplasias Pulmonares/patología , Macrófagos Asociados a Tumores/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Ciclo Celular , Puntos de Control del Ciclo Celular , Movimiento Celular , Técnicas de Cocultivo , Regulación hacia Abajo , Xenoinjertos , Humanos , Inmunoglobulinas/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Células THP-1 , Regulación hacia Arriba
16.
Int J Biol Sci ; 17(7): 1671-1681, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33994852

RESUMEN

The immune checkpoint ligand programmed death-ligand 1 (PD-L1) and the transmembrane mucin (MUC) 3A are upregulated in non-small cell lung cancer (NSCLC), contributing to the aggressive pathogenesis and poor prognosis. Here, we report that knocking down the oncogenic MUC3A suppresses the PD-L1 expression in NSCLC cells. MUC3A is a potent regulator of epidermal growth factor receptor (EGFR) stability, and MUC3A deficiency downregulates the activation of the PI3K/Akt and MAPK pathways, which subsequently reduces the expression of PD-L1. Furthermore, knockdown of MUC3A and tyrosine kinase inhibitors (TKIs) in EGFR-mutant NSCLC cells play a synergistic effect on inhibited proliferation and promoted apoptosis in vitro. In the BALB/c nude mice xenograft model, MUC3A deficiency enhances EGFR-mutated NSCLC sensitivity to TKIs. Our study shows that transmembrane mucin MUC3A induces PD-L1, thereby promoting immune escape in NSCLC, while downregulation of MUC3A enhances TKIs effects in EGFR-mutant NSCLC. These findings offer insights into the design of novel combination treatment for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Mucina 3/genética , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Terapia Molecular Dirigida/métodos , Mucina 3/biosíntesis , Neoplasias Experimentales , Transducción de Señal , Células Tumorales Cultivadas
17.
J Mater Chem B ; 9(23): 4643-4653, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34009230

RESUMEN

Radiotherapy, as well as chemotherapy and surgery, occupies an essential position in tumor treatment. Nonetheless, insufficient radiation deposition and hypoxia-related radioresistance of cancer cells still are serious challenges in radiotherapy. Herein, we proposed a hollow PtCo nanosphere (PtCo NS)-based novel radiosensitizer with three advantages to sensitize tumor radiotherapy: (i) the high-Z element Pt ensured higher radiation absorption to cause more DNA damage, (ii) the platinum (Pt) and cobalt (Co) elements exhibited a dual catalase-like enzymatic activity to convert endogenic H2O2 to O2 efficiently, and (iii) the unique hollow nature of the PtCo NS provided a large specific surface area, which could amplify the catalytic reaction of H2O2 to induce reactive oxygen species and cancer cell apoptosis upon combination with radiation. Both in vivo and in vitro studies showed that the hollow PtCo NS could significantly inhibit tumor growth, simultaneously relieving tumor hypoxia with good biocompatibility and biosafety. This work presents a simple but multifunctional radiosensitizer with a unique hollow structure for radiotherapy enhancement.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Catalasa/metabolismo , Cobalto/química , Neoplasias Pulmonares/radioterapia , Nanopartículas del Metal/química , Nanosferas/química , Oxígeno/metabolismo , Platino (Metal)/química , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Difracción de Polvo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cell Biosci ; 11(1): 74, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33858512

RESUMEN

BACKGROUND: As one of the most common malignancy, lung adenocarcinoma (LUAD) is characterized by low 5-year survival rate. This research aimed to investigate the effects of ribonucleotide reductase regulatory subunit M2 (RRM2) on malignant biological behaviors and activation of cGAS/STING pathway. We also explored the synergistic sensitization mechanisms of RRM2 and radiotherapy. METHODS: Bioinformatic tools were used to evaluate the clinical significance of RRM2 in LUAD patients. The roles of RRM2 in malignant phenotype and DNA damage in LUAD cells were investigated with cell proliferation, colony formation, immunofluorescence, modified Boyden chamber and comet assays. The mouse models were used to evaluate the biological significance of RRM2 in vivo. Cytotoxic T cell infiltration was evaluated via flow cytometric analysis and immunohistochemistry staining in C57BL/6 mice. We also explored the synergistic effects of RRM2 silencing and radiation on LUAD cells with apoptosis assay and immunoblotting in vitro. RESULTS: Bioinformatic analysis revealed that RRM2 had diagnostic values for LUAD patients. Higher levels of RRM2 predicted worse prognosis. RRM2 silencing inhibited LUAD cell proliferation, invasion and migration. RRM2 knockdown induced S phase arrest and DNA damage. RRM2 silencing induced cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway, and the downstream targets were regulated in a STING-dependent manner. Knockdown of RRM2 suppressed tumor growth in the xenograft tumor models. RRM2 deficiency increased CD8 + T cells in the tumor tissues and spleens. Furthermore, RRM2 silencing had synergistic effects with radiation on inhibiting cell proliferation and promoting apoptosis. Meanwhile, this combination promoted the activation of cGAS/STING signaling pathway synergistically, and simultaneously increased expression of IFNß, CCL5 and CXCL10. CONCLUSION: Our results demonstrated that RRM2 silencing had anti-tumor values and activated the cGAS/STING signaling pathway. RRM2 silencing increased CD8 + T cells infiltration. RRM2 silencing cooperated with radiation to inhibit LUAD cell proliferation, promote apoptosis and enhance the activation of cGAS/STING signaling pathway. RRM2 could be a promising target for tumor regression through cancer immunotherapy in LUAD.

19.
Front Oncol ; 11: 804418, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35111682

RESUMEN

Non-small cell lung cancer (NSCLC) has high mortality rates worldwide. Agrin contributes to immune synapse information and is involved in tumor metastasis. However, its roles in NSCLC and tumor immune microenvironment remain unclear. This study examined the effects and the underlying mechanisms of Agrin in NSCLC and tumor-infiltrated immune cells. Clinical tissue samples were used to confirm the bioinformatic predictions. NSCLC cells were used to investigate the effects of Agrin on cell cycle and proliferation, as well as invasion and migration. Tumor xenograft mouse model was used to confirm the effects of Agrin on NSCLC growth and tumor-infiltrated regulatory T cells (Tregs) in vivo. Agrin levels in NSCLC cells were closely related to tumor progression and metastasis, and its function was enriched in the PI3K/AKT pathway. In vitro assays demonstrated that Agrin knockdown suppressed NSCLC cell proliferation and metastasis, while PI3K/AKT activators reversed the inhibitory effects of Agrin deficiency on NSCLC cell behaviors. Agrin expression was negatively associated with immunotherapy responses in NSCLC patients. Agrin knockdown suppressed Tregs, as well as interleukin (IL)-6 expression and secretion, while PI3K/AKT activators and exogenous IL-6 rescued the inhibitory effects. In the mouse model, Agrin downregulation alleviated NSCLC cell growth and Treg infiltration in vivo. Our results indicated that Agrin promotes tumor cell growth and Treg infiltration via increasing IL-6 expression and secretion through PI3K/AKT pathway in NSCLC. Our studies suggested Agrin as a therapeutically potential target to increase the efficacy of immunotherapy in NSCLC patients.

20.
Cancer Med ; 9(24): 9581-9594, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33098370

RESUMEN

Lung adenocarcinoma (LUAD) is the most common and lethal cancer worldwide. Radiotherapy (RT) is widely used at all stages of LUAD, and the development of immunotherapy substantially enhances the survival of LUAD patients. Although the emerging treatments for LUAD have improved prognosis, only a small fraction of patients can benefit from clinical therapies. Thereby, approaches assessing responses to RT and immunotherapy in LUAD patients are essential. After integrating the analysis of RT, immunization, mRNA, and clinical information, we constructed a signature based on 308 tumor-infiltrating B lymphocyte-specific genes (TILBSig) using a machine learning method. TILBSig was composed of 6 B cell-specific genes (PARP15, BIRC3, RUBCNL, SP110, TLE1, and FADS3), which were highly associated with the overall survival as independent factors. TILBSig was able to differentiate better survival compared with worse survival among different patients, and served as an independent factor for clinical characteristics. The low-risk TILBSig group was correlated with more immune cell infiltration (especially B lineages) and lower cancer stem cell characteristics than the high-risk group. The patients with lower risk scores were more likely to respond to RT and immunotherapy. TILBSig served as an excellent predicator for prognosis and response to immunotherapy and RT in LUAD patients.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/terapia , Linfocitos B/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Adenocarcinoma del Pulmón/patología , Anciano , Linfocitos B/patología , Biomarcadores de Tumor/genética , Biología Computacional/métodos , Bases de Datos Genéticas , Femenino , Humanos , Inmunoterapia , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Pronóstico , Radioterapia , Tasa de Supervivencia , Resultado del Tratamiento , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA