Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
BMC Pulm Med ; 24(1): 279, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38867173

RESUMEN

BACKGROUND: Legionella pneumonia is one of the most severe types of atypical pneumonia, impairing multiple organ systems, posing a threat to life. Diagnosing Legionella pneumonia is challenging due to difficulties in culturing the bacteria and limitations in immunoassay sensitivity and specificity. CASE PRESENTATION: This paper reports a rare case of sepsis caused by combined infection with Legionella pneumophila and Fusobacterium necrophorum, leading to respiratory failure, acute kidney injury, acute liver injury, myocardial damage, and electrolyte disorders. In addition, we systematically reviewed literature on patients with combined Legionella infections, analyzing their clinical features, laboratory results and diagnosis. CONCLUSIONS: For pathogens that require prolonged incubation periods and are less sensitive to conventional culturing methods, metagenomic next-generation sequencing (mNGS) can be a powerful supplement to pathogen screening and plays a significant role in the auxiliary diagnosis of complex infectious diseases.


Asunto(s)
Coinfección , Infecciones por Fusobacterium , Fusobacterium necrophorum , Secuenciación de Nucleótidos de Alto Rendimiento , Legionella pneumophila , Enfermedad de los Legionarios , Humanos , Legionella pneumophila/genética , Legionella pneumophila/aislamiento & purificación , Enfermedad de los Legionarios/diagnóstico , Enfermedad de los Legionarios/microbiología , Infecciones por Fusobacterium/diagnóstico , Infecciones por Fusobacterium/microbiología , Infecciones por Fusobacterium/complicaciones , Fusobacterium necrophorum/aislamiento & purificación , Fusobacterium necrophorum/genética , Coinfección/diagnóstico , Coinfección/microbiología , Metagenómica/métodos , Masculino , Persona de Mediana Edad , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/diagnóstico
2.
Comput Biol Med ; 171: 108183, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38422959

RESUMEN

BACKGROUND: As one of the common subtypes of non-small lung cancer, lung squamous cell carcinoma (LUSC) patients with advanced stage have few choices of treatment strategies. Therefore, it is urgent to discover genes that are associated with the survival and efficacy of immunotherapies. METHOD: Differential gene expression analyses were conducted using TCGA LUSC bulk-sequencing and single-cell RNA-sequencing data. Prognostic genes were identified from the TCGA LUSC cohort. Protein expression validation and survival analyses were performed. Experiments were conducted to explore the underlying mechanisms. In addition, the correlation between gene expression and pathological response to adjuvant immunochemotherapy was also investigated. RESULTS: After a series of bioinformatic analyses, solute carrier family 2 member 1(SLC2A1), encoding glucose transporter-1 (GLUT1), was found to be differentially expressed between tumor and normal tissues. GLUT1 was subsequently identified as an independent prognostic factor for LUSC. GSEA analysis revealed the glycolysis metabolism pathway of KEGG enriched in SLC2A1high tumor tissues. LASSO analyses revealed that tumor tissues with high expression of SLC2A1 were associated with high levels of protein lactylation. We found that SLC2A1 was preferentially expressed by SPP1+ macrophages in the tumor microenvironment, and the expression of SLC2A1 was associated with the abundance of SPP1+ macrophages. Immunofluorescence demonstrated GLUT1 and HIF1α colocalization in tumor-infiltrating macrophages. In vitro experiments showed HIF-1α-induced macrophage polarization under hypoxia, and GLUT1 inhibition blocked this polarization. In addition, SLC2A1 was negatively associated with the common immune checkpoint molecules, such as programmed cell death 1(PD-1), T cell immunoreceptor with Ig and ITIM domains (TIGIT), cytotoxic T-lymphocyte associated protein 4 (CTLA4) and lymphocyte activating 3 (LAG3), while showed a positive association with CD44. Finally, we observed that there was a significant correlation between pre-adjuvant-treatment GLUT1 expression and the pathological response. CONCLUSION: SLC2A1 expression was differentially upregulated in tumor tissues, and elevated GLUT1 expression was associated with worse survival and poor pathological response to adjuvant immunochemotherapy. Upregulation of GLUT1 promoted macrophage polarization into the M2 phenotype. The findings will contribute to guiding the treatment selection for LUSC patients and providing personalized immunotherapy strategies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Humanos , Transportador de Glucosa de Tipo 1/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/terapia , Biomarcadores , Inmunoterapia , Pulmón , Microambiente Tumoral
3.
J Cell Physiol ; 233(9): 7447-7456, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29663359

RESUMEN

Non-coding RNAs can exert significant roles various cancers, including NSCLC. Previously, we indicated that lncRNA DGCR5 can promote lung cancer progression through inhibiting hsa-mir-22-3p. In our current study, we investigated the role of DGCR5 in cancer stem cell-like properties of NSCLC. CSCs have been recognized as the frequent cause of tumor metastasis, tumor recurrence, and chemotherapy resistance. Here, lung cancer stem cells were successfully enriched from the parental NSCLC A549, H460, and H1299 cells by using tumor sphere formation assays and side population (SP) assays. We observed that DGCR5 was up-regulated in the enriched CSCs of NSCLC. DGCR5 can inhibit the stemness of NSLCL while overexpression of DGCR5 promoted CSC-like traits. In addition, miR-330-5p was predicted as target of DGCR5 and the correlation between them was validated by dual-luciferase reporter assay, RIP assay, and RNA pull-down assay. Meanwhile, it was found that miR-330-5p was decreased in CSCs of NSCLC. miR-330-5p mimics repressed the stemness while miR-330-5p inhibition enhanced CSC-like properties by targeting CD44. Taken these together, DGCR5 can act as a crucial regulator of CSCs in NSCLC by modulating miR-330-5p/CD44 axis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , ARN Largo no Codificante/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Hialuranos/genética , MicroARNs/genética , Células Madre Neoplásicas/patología , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
J Cell Physiol ; 233(5): 4126-4136, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29030962

RESUMEN

Long non-coding RNAs (lncRNAs) serve critical roles in the pathogenesis of various cancers, including lung adenocarcinoma (LUAD). Herein, in this study, we aimed to investigate the biological and clinical significance of lncRNA DiGeorge syndrome critical region gene 5 (DGCR5) in LUAD. It was observed that DGCR5 was upregulated in LUAD tissues and LUAD cell lines. Inhibition of DGCR5 can prevent LUAD progression via playing anti-apoptosis roles. Both mRNA expression and protein levels of BCL-2 were increased by DGCR5 downregulation while reversely BAX was increased. Additionally, a novel microRNA target of DGCR5, hsa-mir-22-3p was identified through bioinformatics search and confirmed by dual-luciferase reporter system. Gain and loss-of-function studies were performed to verify whether DGCR5 exerts its biological functions through regulating hsa-mir-22-3p in vitro. Overexpression of DGCR5 was able to reverse the tumor inhibitory effect of hsa-mir-22-3p mimics. Furthermore, in vivo tests tumor xenografts were established to detect the function of DGCR5 in LUAD tumorigenesis. Downregulated DGCR5 expression was greatly associated with smaller tumor size, implying a favorable prognosis of LUAD patients. Taken these together, DGCR5 could be considered as a prognostic biomarker and therapeutic target in LUAD diagnosis and treatment.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Biomarcadores de Tumor/genética , Carcinogénesis/genética , ARN Largo no Codificante/genética , Células A549 , Adenocarcinoma del Pulmón/patología , Animales , Apoptosis , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , MicroARNs/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Zhongguo Fei Ai Za Zhi ; 20(2): 114-123, 2017 Feb 20.
Artículo en Chino | MEDLINE | ID: mdl-28228223

RESUMEN

BACKGROUND: A survival analysis and the influencing factors for non-small cell lung cancer (NSCLC) patients with brain metastases accepting first-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKIs) treatment have not yet been elucidated to date. In this study, we collected and analyzed the survival data of NSCLC patients with brain metastasis to obtain evidence and to provide guidance in clinical practice. METHODS: NSCLC patients with brain metastases who were treated with first-generation EGFR-TKIs were retrospectively collected in 2012-2013 from Shanghai Chest Hospital, Shanghai Jiao Tong University. The Kaplan-Meier method and Cox regression were performed for univariate and multivariate analyses, respectively, to explore the independent predictors influencing the survival of patients with NSCLC brain metastases. RESULTS: The median progression-free survival (PFS) and median overall survival (OS) of all patients treated with first-generation EGFR-TKIs were 10.0 months (95%CI: 8.3-11.7) and 28.0 months (95%CI: 22.9-33.1), respectively. Pathological subtypes were the independent predictors of PFS (P=0.001), and tumor differentiations were the independent predictors of OS (P=0.050). CONCLUSIONS: First-generation EGFR-TKIs showed promising efficacy in NSCLC patients with brain metastases. PFS was longer in patients with adenocarcinoma than in those with a non-adenocarcinoma subtype. OS was longer in patients with differentiated tumors than in those who developed poorly differentiated tumors.
.


Asunto(s)
Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Inhibidores de Proteínas Quinasas/uso terapéutico , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...