Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
World J Surg Oncol ; 22(1): 149, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38840197

RESUMO

OBJECTIVE: The purpose of this study was to evaluate the efficacy and clinical value of US, FNAC,FNA-Tg and FNAC + FNA-Tg, as well as the cutoff values of FNA-Tg to evaluate LN metastasis. METHODS: We analyzed the diagnostic value of different US signs, the efficiency of US, FNAC, FNA-Tg and FNAC + FNA-Tg among the LN- and LN + groups, and the cutoff value of FNA-Tg to evaluate LN metastasis. We punctured LNs multiple times and measured the levels of FNA-Tg. Furthermore, the LNs were marked with immunohistochemical Tg and LCA to distinguish the presence of Tg in the para-cancerous tissue of the LNs. RESULTS: The s-Tg and FNA-Tg of the LN + group were higher than those of the LN- group (P = 0.018, ≤ 0.001). The LN + group had more abnormal US signs than the LN- group. The cutoff value of FNA-Tg was 3.2 ng/mL. US had a high sensitivity (92.42), but the specificity was not satisfactory (55.1). FNA-Tg had a higher sensitivity (92.42 vs. 89.39), specificity (100 vs. 93.88), and accuracy (92.42 vs. 83.27) than FNAC. However, the sensitivity of FNAC + FNA-Tg increased further, while the specificity and accuracy decreased slightly. The presence of Tg in the normal lymphocytes adjacent to the cancer was confirmed. CONCLUSION: Ultrasonography provides a noninvasive, dynamic, multidimensional assessment of LNs. With a cutoff value of 3.2 ng/mL, FNA-Tg has higher accuracy and a lower false-negative rate than various single diagnoses. However, FNAC combined with FNA-Tg does not cause additional pain to patients and offers a higher diagnostic efficacy and clinical value.


Assuntos
Metástase Linfática , Tireoglobulina , Neoplasias da Glândula Tireoide , Humanos , Biópsia por Agulha Fina/métodos , Feminino , Metástase Linfática/diagnóstico , Masculino , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/cirurgia , Neoplasias da Glândula Tireoide/diagnóstico , Pessoa de Meia-Idade , Adulto , Tireoglobulina/análise , Tireoglobulina/metabolismo , Prognóstico , Citodiagnóstico/métodos , Carcinoma Papilar/patologia , Carcinoma Papilar/diagnóstico , Carcinoma Papilar/cirurgia , Linfonodos/patologia , Idoso , Seguimentos , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/metabolismo , Ultrassonografia/métodos , Adulto Jovem , Câncer Papilífero da Tireoide/patologia , Câncer Papilífero da Tireoide/cirurgia , Câncer Papilífero da Tireoide/diagnóstico
2.
J Exp Clin Cancer Res ; 43(1): 63, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38424624

RESUMO

BACKGROUND: Lung cancer is one of the most common tumors in the world, and metastasis is one of the major causes of tumor-related death in lung cancer patients. Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and are frequently associated with tumor metastasis in human cancers. However, the regulatory mechanisms of TAMs in lung cancer metastasis remain unclear. METHODS: Single-cell sequencing analysis of lung cancer and normal tissues from public databases and from 14 patients who underwent surgery at Zhongshan Hospital was performed. In vitro co-culture experiments were performed to evaluate the effects of TAMs on lung cancer migration and invasion. Changes in the expression of IL-6, STAT3, C/EBPΒ, and EMT pathway were verified using RT-qPCR, western blotting, and immunofluorescence. Dual luciferase reporter assays and ChIP were used to reveal potential regulatory sites on the transcription factor sets. In addition, the effects of TAMs on lung cancer progression and metastasis were confirmed by in vivo models. RESULTS: TAM infiltration is associated with tumor progression and poor prognosis. IL-6 secreted by TAMs can activate the JAK2/STAT3 pathway through autocrine secretion, and STAT3 acts as a transcription factor to activate the expression of C/EBPß, which further promotes the transcription and expression of IL-6, forming positive feedback loops for IL6-STAT3-C/EBPß-IL6 in TAMs. IL-6 secreted by TAMs promotes lung cancer progression and metastasis in vivo and in vitro by activating the EMT pathway, which can be attenuated by the use of JAK2/STAT3 pathway inhibitors or IL-6 monoclonal antibodies. CONCLUSIONS: Our data suggest that TAMs promote IL-6 expression by forming an IL6-STAT3-C/EBPß-IL6 positive feedback loop. Released IL-6 can induce the EMT pathway in lung cancer to enhance migration, invasion, and metastasis. The use of IL-6-neutralizing antibody can partially counteract the promotion of LUAD by TAMs. A novel mechanism of macrophage-promoted tumor progression was revealed, and the IL6-STAT3-C/EBPß-IL6 signaling cascade may be a potential therapeutic target against lung cancer.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Interleucina-6/metabolismo , Macrófagos Associados a Tumor/metabolismo , Linhagem Celular Tumoral , Retroalimentação , Adenocarcinoma de Pulmão/genética , Neoplasias Pulmonares/genética , Fatores de Transcrição/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Microambiente Tumoral , Transição Epitelial-Mesenquimal
3.
Cancer Lett ; 581: 216497, 2024 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-38008395

RESUMO

Metformin's effect on tumor treatment was complex, because it significantly reduced cancer cell proliferation in vitro, but made no difference in prognosis in several clinical cohorts. Our transcriptome sequencing results revealed that tumor-associated macrophage (TAM) infiltration significantly increased in active lung adenocarcinoma (LUAD) patients with long-term metformin use. We further identified that the tumor suppressive effect of metformin was more significant in mice after the depletion of macrophages, suggesting that TAMs might play an important role in metformin's effects in LUAD. Combining 10X Genomics single-cell sequencing of tumor samples, transcriptome sequencing of metformin-treated TAMs, and the ChIP-Seq data of the Encode database, we identified and validated that metformin significantly increased the expression and secretion of S100A9 of TAMs through AMPK-CEBP/ß pathway. For the downstream, S100A9 binds to RAGE receptors on the surface of LUAD cells, and then activates the NF-κB pathway to promote EMT and progression of LUAD, counteracting the inhibitory effect of metformin on LUAD cells. In cell-derived xenograft models (CDX) and patient-derived xenograft models (PDX) models, our results showed that neutralizing antibodies targeting TAM-secreted S100A9 effectively enhanced the tumor suppressive effect of metformin in treating LUAD. Our results will enable us to better comprehend the complex role of metformin in LUAD, and advance its clinical application in cancer treatment.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Metformina , Animais , Humanos , Camundongos , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/metabolismo , Calgranulina B/genética , Modelos Animais de Doenças , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Metformina/farmacologia , Metformina/uso terapêutico , Prognóstico , Macrófagos Associados a Tumor/metabolismo
5.
Cell Biosci ; 13(1): 103, 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37291676

RESUMO

BACKGROUND: Neoadjuvant chemotherapy (NACT) becomes the first-line option for advanced tumors, while patients who are not sensitive to it may not benefit. Therefore, it is important to screen patients suitable for NACT. METHODS: Single-cell data of lung adenocarcinoma (LUAD) and esophageal squamous carcinoma (ESCC) before and after cisplatin-containing (CDDP) NACT and cisplatin IC50 data of tumor cell lines were analyzed to establish a CDDP neoadjuvant chemotherapy score (NCS). Differential analysis, GO, KEGG, GSVA and logistic regression models were performed by R. Survival analysis were applied to public databases. siRNA knockdown in A549, PC9, TE1 cell lines, qRT-PCR, western-blot, cck8 and EdU experiments were used for further verification in vitro. RESULTS: 485 genes were expressed differentially in tumor cells before and after neoadjuvant treatment for LUAD and ESCC. After combining the CDDP-associated genes, 12 genes, CAV2, PHLDA1, DUSP23, VDAC3, DSG2, SPINT2, SPATS2L, IGFBP3, CD9, ALCAM, PRSS23, PERP, were obtained and formed the NCS score. The higher the score, the more sensitive the patients were to CDDP-NACT. The NCS divided LUAD and ESCC into two groups. Based on differentially expressed genes, a model was constructed to predict the high and low NCS. CAV2, PHLDA1, ALCAM, CD9, IGBP3 and VDAC3 were significantly associated with prognosis. Finally, we demonstrated that the knockdown of CAV2, PHLDA1 and VDAC3 in A549, PC9 and TE1 significantly increased the sensitivity to cisplatin. CONCLUSIONS: NCS scores and related predictive models for CDDP-NACT were developed and validated to assist in selecting patients who might benefit from it.

6.
PeerJ ; 11: e15377, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37180584

RESUMO

Background: Lung adenocarcinoma is one of the most prevalent cancers while ferroptosis is crucial for cancer therapies. This study aims to investigate the function and mechanism of hepatic nuclear factor 4 alpha (HNF4A) in lung adenocarcinomas' ferroptosis. Materials and Methods: HNF4A expression in ferroptotic A549 cells was detected. Then HNF4A was knocked down in A549 cells while overexpressed in H23 cells. Cells with changed HNF4A expression were tested for cytotoxicity and the level of cellular lipid peroxidation. The expression of cytochrome P450 oxidoreductase (POR) expression was examined after HNF4A was knocked down or overexpressed. Chromatin immunoprecipitation-quantitative PCR (ChIP-qPCR) and dual-luciferase assays were performed to validate the regulation of HNF4A on POR. Finally, POR was restored in HNF4A-altered cells to check whether it restores the effect of HNF4A on ferroptosis. Results: We found that HNF4A expression significantly decreased in the ferroptosis of A549 cells, and this change can be blocked by deferoxamine, an inhibitor of ferroptosis. Knockdown of HNF4A inhibited ferroptosis in A549 cells while overexpression of HNF4A promoted ferroptosis in H23 cells. We identified a key ferroptosis-related gene, POR serves as a potential target gene of HNF4A, whose expression was significantly changed in lung adenocarcinoma cells knocking down or overexpressing HNF4A. We demonstrated that HNF4A was bound to the POR's promoter to enhance POR expression, and identified the binding sites via ChIP-qPCR and luciferase assays. Restoration of POR expression blocked the promoting effect of HNF4A on ferroptosis in lung adenocarcinoma. Conclusion: HNF4A promotes POR expression through binding to the POR's promoter, and subsequently promotes the ferroptosis of lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão , Ferroptose , Neoplasias Pulmonares , Humanos , Ferroptose/genética , Ativação Transcricional , Sistema Enzimático do Citocromo P-450/metabolismo , Adenocarcinoma de Pulmão/genética , Neoplasias Pulmonares/genética , Luciferases/metabolismo
7.
Endocrinology ; 163(9)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35881915

RESUMO

OBJECTIVE: Abnormal expression of CUT-like homeobox 2 gene (CUX2) has been highlighted as potential clinical biomarkers in human cancers. Notably, the function of CUX2 has been less elucidated in breast cancer (BC). We focused on the role of the CUX2 in tumorigenesis and progression of BC with the involvement of the lysine demethylase 5B (KDM5B)/sex determining region Y-box 17 (SOX17) axis. METHODS: CUX2, KDM5B, and SOX17 expression levels in BC tissues and cells were tested by reverse transcription quantitative PCR and Western blotting. Later, the effects of CUX2, KDM5B, and SOX17 on the malignant behaviors of MDA-MB-231 and MCF-7 cells were analyzed by CCK-8, colony formation, and Transwell assays in vitro. The interactions of CUX2, KDM5B, and SOX17 were validated by online website prediction, ChIP assay, and dual luciferase reporter gene assay. The subcutaneous tumorigenesis in nude mice was conducted to observe the roles of CUX2, KDM5B, and SOX17 in BC tumor growth in vivo. RESULTS: CUX2 and KDM5B were highly expressed while SOX17 had low expression in BC. Inhibition of CUX2 suppressed BC cell malignant phenotypes. CUX2 promoted KDM5B expression through transcriptional activation, enabling its high expression in BC. KDM5B inhibited SOX17 expression through histone demethylation. Overexpression of KDM5B or downregulation of SOX17 reversed the inhibitory effect of CUX2 downregulation on the malignant behaviors of BC cells. Inhibition of CUX2 impeded BC cell growth in vivo through the KDM5B/SOX17 axis. CONCLUSION: This study highlights that suppression of CUX2 inhibits KDM5B to repress tumorigenesis and progression of BC through overexpressing SOX17.


Assuntos
Neoplasias da Mama , MicroRNAs , Animais , Neoplasias da Mama/metabolismo , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas HMGB , Proteínas de Homeodomínio , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos , Camundongos Nus , MicroRNAs/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo
8.
J Transl Med ; 20(1): 171, 2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35410350

RESUMO

OBJECTIVES: Platinum-based chemotherapies are currently the first-line treatment of non-small cell lung cancer. This study will improve our understanding of the causes of resistance to cisplatin, especially in lung adenocarcinoma (LUAD) and provide a reference for therapeutic decisions in clinical practice. METHODS: Cancer Cell Line Encyclopedia (CCLE), The Cancer Genome Atlas (TCGA) and Zhongshan hospital affiliated to Fudan University (zs-cohort) were used to identify the multi-omics differences related to platinum chemotherapy. Cisplatin-resistant mRNA and miRNA models were constructed by Logistic regression, classification and regression tree and C4.5 decision tree classification algorithm with previous feature selection performed via least absolute shrinkage and selection operator (LASSO). qRT-PCR and western-blotting of A549 and H358 cells, as well as single-cell Seq data of tumor samples were applied to verify the tendency of certain genes. RESULTS: 661 cell lines were divided into three groups according to the IC50 value of cisplatin, and the top 1/3 (220) with a small IC50 value were defined as the sensitive group while the last 1/3 (220) were enrolled in the insensitive group. TP53 was the most common mutation in the insensitive group, in contrast to TTN in the sensitive group. 1348 mRNA, 80 miRNA, and 15 metabolites were differentially expressed between 2 groups (P < 0.05). According to the LASSO penalized logistic modeling, 6 of the 1348 mRNAs, FOXA2, BATF3, SIX1, HOXA1, ZBTB38, IRF5, were selected as the associated features with cisplatin resistance and for the contribution of predictive mRNA model (all of adjusted P-values < 0.001). Three of 6 (BATF3, IRF5, ZBTB38) genes were finally verified in cell level and patients in zs-cohort. CONCLUSIONS: Somatic mutations, mRNA expressions, miRNA expressions, metabolites and methylation were related to the resistance of cisplatin. The models we created could help in the prediction of the reaction and prognosis of patients given platinum-based chemotherapies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Proteínas de Homeodomínio , Humanos , Fatores Reguladores de Interferon , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Aprendizado de Máquina , MicroRNAs/genética , RNA Mensageiro/genética , RNA Mensageiro/uso terapêutico
9.
Int J Biol Sci ; 18(2): 522-535, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35002507

RESUMO

Background: Lung adenocarcinoma (LUAD), the major subtype of lung cancer, is among the leading cause of cancer-related death worldwide. Energy-related metabolic reprogramming metabolism is a hallmark of cancer shared by numerous cancer types, including LUAD. Nevertheless, the functional pathways and molecular mechanism by which FAM83A-AS1 acts in metabolic reprogramming in lung adenocarcinoma have not been fully elucidated. Methods: We used transwell, wound-healing scratch assay, and metabolic assays to explore the effect of FAM83A-AS1 in LUAD cell lines. Western blotting, Co-IP assays, and ubiquitination assays were used to detect the effects of FAM83A-AS1 on HIF-1α expression, degradation, and its binding to VHL. Moreover, an in vivo subcutaneous tumor formation assay was used to detect the effect of FAM83A-AS1 on LUAD. Results: Herein, we identified FAM83A-AS1 as a metabolism-related lncRNA, which was highly correlated with glycolysis, hypoxia, and OXPHOS pathways in LUAD patients using bioinformatics analysis. In addition, we uncovered that FAM83A-AS1 could promote the migration and invasion of LUAD cells, as well as influence the stemness of LUAD cells in vivo and vitro. Moreover, FAM83A-AS1 was shown to promote glycolysis in LUAD cell lines in vitro and in vivo, and was found to influence the expression of genes related to glucose metabolism. Besides, we revealed that FAM83A-AS1 could affect glycolysis by regulating HIF-1α degradation. Finally, we found that FAM83A-AS1 knockdown could inhibit tumor growth and suppress the expression of HIF-1α and glycolysis-related genes in vivo. Conclusion: Our study demonstrates that FAM83A-AS1 contributes to LUAD proliferation and stemness via the HIF-1α/glycolysis axis, making it a potential biomarker and therapeutic target in LUAD patients.


Assuntos
Adenocarcinoma de Pulmão/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , RNA Longo não Codificante/genética , Efeito Warburg em Oncologia , Células A549 , Adenocarcinoma de Pulmão/patologia , Animais , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Immunol Immunother ; 71(7): 1733-1746, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34837101

RESUMO

Metformin has been found to have inhibitory effects on a variety of tumors. However, its effects on non-small cell lung cancer (NSCLC) remain unclear. We demonstrated that metformin could inhibit the proliferation of A549 and H1299 cells. RNA transcriptome sequencing revealed that PDL1 was significantly downregulated in both cell types following treatment with metformin (P < 0.001). Jaspar analysis and chromatin immunoprecipitation showed that CEBPB could directly bind the promoter region of PDL1. Western blotting showed that protein expression of the isoforms CEBPB-LAP*, CEBPB-LAP, and CEBPB-LIP was significantly upregulated and the LIP/LAP ratio was increased. Gene chip analysis showed that PDL1 was significantly upregulated in A549-CEBPB-LAP cells and significantly downregulated in A549-CEBPB-LIP cells (P < 0.05) compared with CEBPB-NC cells. Dual-luciferase reporter gene assay showed that CEBPB-LAP overexpression could promote transcription of PDL1 and CEBPB-LIP overexpression could inhibit the process. Functional assays showed that the changes in CEBPB isoforms affected the function of NSCLC cells. Western blotting showed that metformin could regulate the function of NSCLC cells via AMPK-CEBPB-PDL1 signaling. Animal experiments showed that tumor growth was significantly inhibited by metformin, and atezolizumab and metformin had a synergistic effect on tumor growth. A total of 1247 patients were retrospectively analyzed, including 166 and 1081 patients in metformin and control groups, respectively. The positive rate of PDL1 was lower than that of the control group (HR = 0.338, 95% CI = 0.235-0.487; P < 0.001). In conclusion, metformin inhibited the proliferation of NSCLC cells and played an anti-tumor role in an AMPK-CEBPB-PDL1 signaling-dependent manner.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Metformina , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/farmacologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Metformina/farmacologia , Metformina/uso terapêutico , Estudos Retrospectivos , Transdução de Sinais
11.
J Exp Clin Cancer Res ; 40(1): 398, 2021 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930376

RESUMO

BACKGROUND: Most cancer cells have fundamentally different metabolic characteristics, particularly much higher glycolysis rates than normal tissues, which support the increased demand for biosynthesis and promote tumor progression. We found that transforming growth factor (TGF)-ß plays a dual function in regulating glycolysis and cell proliferation in non-small cell lung cancer. METHODS: We used the PET/MRI imaging system to observe the glucose metabolism of subcutaneous tumors in nude mice. Energy metabolism of non-small cell lung cancer cell lines detected by the Seahorse XFe96 cell outflow analyzer. Co-immunoprecipitation assays were used to detect the binding of Smads and HIF-1α. Western blotting and qRT-PCR were used to detect the regulatory effects of TGF-ß and HIF-1α on c-MYC, PKM1/2, and cell cycle-related genes. RESULTS: We discovered that TGF-ß could inhibit glycolysis under normoxia while significantly promoting tumor cells' glycolysis under hypoxia in vitro and in vivo. The binding of hypoxia-inducible factor (HIF)-1α to the MH2 domain of phosphorylated Smad3 switched TGF-ß function to glycolysis by changing Smad partners under hypoxia. The Smad-p107-E2F4/5 complex that initially inhibited c-Myc expression was transformed into a Smad-HIF-1α complex that promoted the expression of c-Myc. The increased expression of c-Myc promoted alternative splicing of PKM to PKM2, resulting in the metabolic reprogramming of tumor cells. In addition, the TGF-ß/Smad signal lost its effect on cell cycle regulatory protein p15/p21. Furthermore, high expression of c-Myc inhibited p15/p21 and promoted the proliferation of tumor cells under hypoxia. CONCLUSIONS: Our results indicated that HIF-1α functions as a critical factor in the dual role of TGF-ß in tumor cells, and may be used as a biomarker or therapeutic target for TGF-ß mediated cancer progression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Glucose/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares/genética , Imageamento por Ressonância Magnética/métodos , Tomografia por Emissão de Pósitrons/métodos , Fator de Crescimento Transformador beta1/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células , Reprogramação Celular , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Transdução de Sinais , Transfecção
12.
BMC Pulm Med ; 21(1): 316, 2021 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-34635074

RESUMO

BACKGROUND: Differences in genetics and microenvironment of LUAD patients with or without TP53 mutation were analyzed to illustrate the role of TP53 mutation within the carcinogenesis of LUAD, which will provide new concepts for the treatment of LUAD. METHODS: In this study, we used genetics and clinical info from the TCGA database, including somatic mutations data, RNA-seq, miRNA-seq, and clinical data. More than one bioinformatics tools were used to analyze the unique genomic pattern of TP53-related LUAD. RESULTS: According to TP53 gene mutation status, we divided the LUAD patients into two groups, including 265 in the mutant group (MU) and 295 in the wild-type group (WT). 787 significant somatic mutations were detected between the groups, including mutations in titin (TTN), type 2 ryanodine receptor (RYR2) and CUB and Sushi multiple domains 3(CSMD3), which were up-regulated in the MU. However, no significant survival difference was observed. At the RNA level, we obtained 923 significantly differentially expressed genes; in the MU, α-defensin 5(DEFA5), pregnancy-specific glycoprotein 5(PSG5) and neuropeptide Y(NPY) were the most up-regulated genes, glucose-6-phosphatase (G6PC), alpha-fetoprotein (AFP) and carry gametocidal (GC) were the most down-regulated genes. GSVA analysis revealed 30 significant pathways. Compared with the WT, the expression of 12 pathways in the mutant group was up-regulated, most of which pointed to cell division. There were significant differences in tumor immune infiltrating cells, such as Macrophages M1, T cells CD4 memory activated, Mast cells resting, and Dendritic cells resting. In terms of immune genes, a total of 35 immune-related genes were screened, of which VGF (VGF nerve growth factor inducible) and PGC (peroxisome proliferator-activated receptor gamma coactivator) were the most significant up-regulated and down-regulated genes, respectively. Research on the expression pattern of immunomodulators found that 9 immune checkpoint molecules and 6 immune costimulatory molecules were considerably wholly different between the two groups. CONCLUSIONS: Taking the mutant group as a reference, LUAD patients in the mutant group had significant differences in somatic mutations, mRNA-seq, miRNA-seq, immune infiltration, and immunomodulators, indicating that TP53 mutation plays a crucial role in the occurrence and development of LUAD.


Assuntos
Adenocarcinoma de Pulmão/genética , Neoplasias Pulmonares/genética , Microambiente Tumoral , Proteína Supressora de Tumor p53/genética , Idoso , Bases de Dados Factuais , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Checkpoint Imunológico/genética , Masculino , Pessoa de Meia-Idade , Mutação , RNA-Seq
13.
Oncogenesis ; 10(10): 71, 2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34697289

RESUMO

Esophageal squamous cell carcinoma (ESCC) accounts for 90% of all cases of esophageal cancers worldwide. Although neoadjuvant chemotherapy (NACT-ESCC) improves the survival of ESCC patients, the five-year survival rate of these patients is dismal. The tumor microenvironment (TME) and tumor heterogeneity decrease the efficacy of ESCC therapy. In our study, 113,581 cells obtained from five ESCC patients who underwent surgery alone (SA-ESCC) and five patients who underwent preoperative paclitaxel plus platinum chemotherapy (NACT-ESCC), were used for scRNA-seq analysis to explore molecular and cellular reprogramming patterns. The results showed samples from NACT-ESCC patients exhibited the characteristics of malignant cells and TME unlike samples from SA-ESCC patients. Cancer cells from NACT-ESCC samples were mainly at the 'intermediate transient stage'. Stromal cell dynamics showed molecular and functional shifts that formed the immune-activation microenvironment. APOE, APOC1, and SPP1 were highly expressed in tumor-associated macrophages resulting in anti-inflammatory macrophage phenotypes. Levels of CD8+ T cells between SA-ESCC and NACT-ESCC tissues were significantly different. Immune checkpoints analysis revealed that LAG3 is a potential immunotherapeutic target for both NACT-ESCC and SA-ESCC patients. Cell-cell interactions analysis showed the complex cell-cell communication networks in the TME. In summary, our findings elucidate on the molecular and cellular reprogramming of NACT-ESCC and ESCC patients. These findings provide information on the potential diagnostic and therapeutic targets for ESCC patients.

14.
Clin Lung Cancer ; 22(4): 324-331, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33789831

RESUMO

BACKGROUND: We aimed to validate the clinicopathologic characteristics and prognostic value of the presence of solid components in the mediastinal window of computed tomography scan in clinical stage I pulmonary subsolid nodules (SSNs). METHODS: We retrospectively evaluated patients with pulmonary SSNs resected between 2011 and 2016. We classified SSNs into heterogeneous ground-glass nodules (HGGNs) (solid component detected only in lung window) and part-solid nodules (PSNs) (solid component detected both in lung/mediastinal windows). RESULTS: A total of 487 patients (216 PSNs) were included. PSNs were associated with higher frequencies of micropapillary or solid pathologic patterns (18.1% vs. 3.3%; P < .001), epidermal growth factor receptor gene mutation (39.4% vs. 32.8%), and other types of gene mutations (2.3% vs. 1.1%; P = .043). Logistic regression analysis revealed that male sex (odds ratio [OR], 2.58; 95% confidence interval [CI], 1.20-5.57; P = .016) and higher consolidation tumor ratio (CTR) (OR, 110.04; 95% CI, 8.56-1414.39; P < .001) remained independent for invasive adenocarcinomas with poor differentiation. Receiver operating characteristic analyses revealed that solid component size in the mediastinal window (area under the curve [AUC], 0.731; 95% CI, 0.653-0.808; P < .0001) showed a better predictive ability to poor differentiation compared with solid component size in the lung window and CTR. The 5-year recurrence-free survival (RFS) rate of PSNs was worse than that of HGGNs (94.6% vs. 99.1%; P = .019). Multivariate Cox regression revealed that positive lymph node status (hazard ratio, 22.99; 95% CI, 4.52-116.86; P < .001) indicated worse RFS for PSNs. CONCLUSION: SSNs with solid components in mediastinal window demonstrated clinicopathologic and prognostic features different from those without in clinical stage I lung cancer. Solid components in mediastinal window was a strong predictor of poor differentiation.


Assuntos
Adenocarcinoma de Pulmão/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Idoso , Intervalo Livre de Doença , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos , Fatores Sexuais
15.
Front Oncol ; 11: 616149, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33912445

RESUMO

OBJECTIVES: Our study aimed to validate pathologic findings of ground-glass nodules (GGOs) of different consolidation tumor ratios (CTRs), and to explore whether GGOs could be stratified according to CTR with an increment of 0.25 based on its prognostic role. METHODS: We retrospectively evaluated patients with clinical stage IA GGOs who underwent curative resection between 2011 and 2016. The patients were divided into 4 groups according to CTR step by 0.25. Cumulative survival rates were calculated by the Kaplan-Meier method. Univariate and multivariate Cox regression analyses were conducted to obtain the risk factors on relapse-free survival (RFS). The surv_function of the R package survminer was used to determine the optimal cutoff value. Receiver operating characteristic (ROC) analysis was generated to validate optimal cutoff points of factors. RESULTS: A total of 862 patients (608 women; median age, 59y) were included, with 442 patients in group A (CTR ≤ 0.25), 210 patients in group B (0.25

16.
Transl Cancer Res ; 10(11): 4727-4738, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35116327

RESUMO

BACKGROUND: Lung adenocarcinoma (ADC) at stage IB has its own prognostic characteristics. This study aimed to investigate the clinical factors that may affect the prognosis of patients with stage IB ADC. METHODS: The data of ADC cases were selected from the Surveillance, Epidemiology, and End Results (SEER) database (2010-2016) and patients in Zhongshan Hospital, Fudan University (Department of Thoracic Surgery, 2015-2016). Kaplan-Meier method was used to obtain the overall survival (OS). Factors that significantly related to the prognosis were evaluated by univariate and multivariate analysis (UVA, MVA) using the Cox model. A nomogram was developed and validated to predict the 3-year OSs of those patients. RESULTS: 7,605 patients with stage IB ADC were included ultimately and were divided into two groups, a training cohort (n=5,324) and a test cohort (n=2,281). Besides, there was a validation cohort (n=272) for the verification of the nomogram model. Those with significantly older age, male, the white race, lower grades of tumor differentiation, larger tumor size (31-40 mm) without pleural layer (PL) invasion as well as receiving sublobectomy suffered from poorer survival (P<0.001), which were identified as independent factors for stage IB ADC (P<0.001), and according to which, a nomogram model was created. CONCLUSIONS: Age, sex, race, histological grade, surgery to the primary site, and tumor size combined with PL invasion were independent risk factors for stage IB ADC, based on which a nomogram was constructed to predict the prognosis.

17.
Cancer Imaging ; 20(1): 87, 2020 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-33308323

RESUMO

BACKGROUND: The computed tomography (CT) characteristic of ground glass opacity (GGO) were shown to be associated with clinical significance in lung adenocarcinoma. We evaluated the prognostic value of the solid component ratio of GGO IA invasive lung adenocarcinoma. METHODS: We retrospectively analyzed the records of GGO IA patients who received surgical resection from April 2012 to December 2015. The solid component ratio was calculated based on thin-slice CT scans. Baseline features were compared stratified by the ratio. Cox proportional hazard models and survival analyses were adopted to explore potential prognostic value regarding overall survival (OS) and disease-free survival (DFS). RESULTS: Four hundred fifteen patients were included. The higher ratio was significantly associated with larger tumor diameter, pathological subtypes and choice of surgical type. There was a significantly worse DFS with a > 50% ratio. The subgroups of 0% and ≤ 50% ratio showed close survival curves of DFS. Similar trends were observed in OS. Multivariate analyses revealed that the ratio was a significant predictor for DFS, but not for OS. No significant prognostic difference was observed between lobectomy and limited resections. CONCLUSION: A higher solid component ratio may help to predict a significantly worse prognosis of GGO IA lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Adenocarcinoma de Pulmão/mortalidade , Adenocarcinoma de Pulmão/patologia , Adulto , Idoso , Intervalo Livre de Doença , Feminino , Humanos , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos
18.
Cell Discov ; 6: 69, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33083004

RESUMO

As an early type of lung adenocarcinoma, ground glass nodule (GGN) has been detected increasingly and now accounts for most lung cancer outpatients. GGN has a satisfactory prognosis and its characteristics are quite different from solid adenocarcinoma (SADC). We compared the GGN adenocarcinoma (GGN-ADC) with SADC using the single-cell RNA sequencing (scRNA-seq) to fully understand GGNs. The tumor samples of five patients with lung GGN-ADCs and five with SADCs underwent surgery were digested to a single-cell suspension and analyzed using 10× Genomic scRNA-seq techniques. We obtained 60,459 cells and then classified them as eight cell types, including cancer cells, endothelial cells, fibroblasts, T cells, B cells, Nature killer cells, mast cells, and myeloid cells. We provided a comprehensive description of the cancer cells and stromal cells. We found that the signaling pathways related to cell proliferation were downregulated in GGN-ADC cancer cells, and stromal cells had different effects in GGN-ADC and SADC based on the analyses of scRNA-seq results. In GGN-ADC, the signaling pathways of angiogenesis were downregulated, fibroblasts expressed low levels of some collagens, and immune cells were more activated. Furthermore, we used flow cytometry to isolate the cancer cells and T cells in 12 GGN-ADC samples and in an equal number of SADC samples, including CD4+ T and CD8+ T cells, and validated the expression of key molecules by quantitative real-time polymerase chain reaction analyses. Through comprehensive analyses of cell phenotypes in GGNs, we provide deep insights into lung carcinogenesis that will be beneficial in lung cancer prevention and therapy.

19.
Transl Lung Cancer Res ; 9(4): 1407-1421, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32953513

RESUMO

BACKGROUND: Non-smoking-related lung adenocarcinoma (LUAD) has its own characteristics. Genetic and microenvironmental differences in smoking and non-smoking LUAD patients were analyzed to elucidate the oncogenesis of non-smoking-related LUAD, which will improve our understanding of the underlying molecular mechanism and be of clinical use in the future. METHODS: The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) databases were used for clinical and genomic information. Various bioinformatics tools were used to analyze differences in somatic mutations, RNA and microRNA (miRNA) expression, immune infiltration, and stemness indices. GO, KEGG, and GSVA analyses were performed with R. A merged protein-protein interaction (PPI) network was constructed and analyzed. A miRNA-differentially expressed gene network was constructed with miRNet. qRT-PCR was used for validation of 4 most significantly differently expressed genes and 2 miRNAs in tumor samples obtained from 20 pairs of non-smoking and smoking patients. RESULTS: Five hundred and one patients with LUAD were obtained, including 210 in the non-smoking group and 292 in the smoking group. A total of 174 significantly altered somatic mutations were detected, including mutations in tumor protein p53 and epidermal growth factor receptor, which were downregulated in non-smoking-related LUAD. At the RNA level, 231 significantly differentially expressed genes were obtained; 124 were upregulated and 107 downregulated in the non-smoking group. GSVA analysis revealed 42 significant pathways. Other functional and enrichment analyses of somatic mutations and RNA expression levels revealed that these genes were significantly enriched in receptor activity regulation and receptor binding. Differences in microenvironments including immune infiltration (e.g., CD8+ T cells and resting mast cells) and stemness indices were also found between groups. A 79-pair interaction was found between differentially expressed genes and miRNAs, of which miR-335-5p and miR-34a-5p were located in the center. Twenty-one genes, including vitronectin, neurotensin, and neuronatin, were differentially expressed in both non-smoking LUAD patients and DMSO-treated A549 cells. And the different expression of neurotensin, neuronatin, trefoil factor family2, regenerating family member 4, miR-377-5p, miR-34a were verified with the same tendency in our own samples. CONCLUSIONS: Non-smoking LUAD patients, compared to smokers, have different characteristics in terms of somatic mutation, gene, and miRNA expression and the microenvironment, indicating a diverse mechanism of oncogenesis.

20.
Int Immunopharmacol ; 88: 106910, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32829091

RESUMO

BACKGROUND: Immunotherapy has achieved excellent results in patients with lung squamous cell carcinoma. However, in which population it can exert the greatest effect is still unknown. Some studies have suggested that its effect is related to the expression level of PD1. Analyzing the relationship between PD1 expression level and genetic differences in lung squamous cell carcinoma patients will be helpful in understanding the underlying causes of this immunotherapy effect and provide a reference for clinical practice. METHODS: In this study, we used RNA-seq, miRNA-seq, methylation array, mutation profiles, and copy number variation data from the TCGA database and RNA-seq data from the GEO database to analyze the distinctive genomic patterns associated with PD1 and PDL1 expression. RNA-seq data from 44 LUSC patients who underwent surgery at Zhongshan Hospital were also included in the study. RESULTS: After grouping LUSC patients according to the expression levels of PD1 and PDL1, we found no significant difference in survival between the two groups. However, 178 genes, including IL-21, KLRC3, and KLRC4, were significantly upregulated in both the TCGA and GEO databases in the high expression group, and there was a precise correlation between gene expression and epigenetic changes in the two groups. At the same time, the overall level of somatic mutations was not significantly different between the two groups. It is worth noting that the gene enrichment results showed that the differential pathways were mainly enriched in immune regulation, especially T cell-related immune activities. CONCLUSION: We found that the differences in gene expression between the two groups were related to immunity, which may affect the effectiveness of immunotherapy. We hope our results can provide a reference for further research and help in finding other targets to improve the efficacy of immunotherapy.


Assuntos
Antígeno B7-H1/genética , Carcinoma de Células Escamosas/genética , Neoplasias Pulmonares/genética , Receptor de Morte Celular Programada 1/genética , Idoso , Variações do Número de Cópias de DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Genômica , Humanos , Masculino , MicroRNAs , Pessoa de Meia-Idade , Mutação , RNA-Seq
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA