Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
2.
BMC Cancer ; 20(1): 682, 2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32698787

ABSTRACT

BACKGROUND: Long intergenic non-coding RNA 00511 (LINC00511) is highly expressed in diverse cancers and has a correlation with poor clinical outcomes for cancer patients. In view of contradictory data among published data, we aim to evaluate the prognostic role of LINC00511 for cancer patients. METHODS: In the present study, a meta-analysis of related studies has been performed to investigate the prognostic significance of LINC00511 in cancer patients. Relevant studies published before December 22, 2019 were systematically searched online in PubMed, EMBASE, Web of Science, and the Cochrane Library databases. The relationship between LINC00511 expression and cancer patients' survival, including overall survival (OS), disease-free survival (DFS)/relapse-free survival (RFS) and progression-free survival (PFS), was evaluated using pooled hazard ratios (HRs) with their corresponding 95% confidence intervals (CIs). The association between LINC00511 expression and clinicopathological features was assessed using odd ratios (ORs) and their corresponding 95% CIs. RESULTS: A total of 14 eligible studies with 1883 patients were enrolled in the present meta-analysis. The results demonstrated that elevated expression of LINC00511 was significantly associated with poor OS (HR = 2.62; 95% CI: 2.00-3.45; p <  0.001), PFS (HR = 1.80; 95% CI: 1.29-2.51; p = 0.001) and DFS/RFS (HR = 2.90; 95% CI: 1.04-8.12; p = 0.04). Additionally, High LINC00511 expression was associated with large tumor size (OR = 3.10; 95% CI: 1.97-4.86; p <  0.00001), lymph node metastasis (OR = 3.11; 95% CI: 2.30-4.21; p <  0.00001), advanced clinical stage (OR = 3.95; 95% CI: 2.68-5.81; p <  0.00001), distant metastasis (OR = 2.39; 95% CI: 1.16-4.93; p = 0.02), and disease recurrence (OR = 4.62; 95% CI: 2.47-8.65; p <  0.00001). Meanwhile, no correlation was found between LINC00511 expression and age, gender, and histological grade. These findings were consolidated by the results of bioinformatics analysis. CONCLUSIONS: Based on our findings, LINC00511 may serve as a novel prognostic biomarker for cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Neoplasms/mortality , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , Humans , Prognosis , Publication Bias
3.
Lancet Infect Dis ; 20(12): 1457-1469, 2020 12.
Article in English | MEDLINE | ID: mdl-32673595

ABSTRACT

BACKGROUND: Use of an interferon-γ (IFN-γ) release assay or tuberculin skin test for detection and management of latent tuberculosis infection is controversial. For both types of test, we assessed their predictive value for the progression of latent infection to active tuberculosis disease, the targeting value of preventive treatment, and the necessity of dual testing. METHODS: In this systematic review and meta-analysis, we searched PubMed, Embase, Web of Science, and the Cochrane Library, with no start date or language restrictions, on Oct 18, 2019, using the keywords ("latent tuberculosis" OR "latent tuberculosis infection" OR "LTBI") AND ("interferon gamma release assays" OR "Interferon-gamma Release Test" OR "IGRA" OR "QuantiFERON®-TB in tube" OR "QFT" OR "T-SPOT.TB") AND ("tuberculin skin test" OR "tuberculin test" OR "Mantoux test" OR "TST"). We included articles that used a cohort study design; included information that individuals with latent tuberculosis infection detected by IFN-γ release assay, tuberculin skin test, or both, progressed to active tuberculosis; reported information about treatment; and were limited to high-risk populations. We excluded studies that included patients with active or suspected tuberculosis at baseline, evaluated a non-commercial IFN-γ release assay, and had follow-up of less than 1 year. We extracted study details (study design, population investigated, tests used, follow-up period) and the number of individuals observed at baseline, who progressed to active tuberculosis, and who were treated. We then calculated the pooled risk ratio (RR) for disease progression, positive predictive value (PPV), and negative predictive value (NPV) of IFN-γ release assay versus tuberculin skin test. FINDINGS: We identified 1823 potentially eligible studies after exclusion of duplicates, of which 256 were eligible for full-text screening. From this screening, 40 studies (50 592 individuals in 41 cohorts) were identified as eligible and included in our meta-analysis. Pooled RR for the rate of disease progression in untreated individuals who were positive by IFN-γ release assay versus those were negative was 9·35 (95% CI 6·48-13·49) compared with 4·24 (3·30-5·46) for tuberculin skin test. Pooled PPV for IFN-γ release assay was 4·5% (95% CI 3·3-5·8) compared with 2·3% (1·5-3·1) for tuberculin skin test. Pooled NPV for IFN-γ release assay was 99·7% (99·5-99·8) compared with 99·3% (99·0-99·5) for tuberculin skin test. Pooled RR for rates of disease progression in individuals positive by IFN-γ release assay who were untreated versus those who were treated was 3·09 (95% CI 2·08-4·60) compared with 1·11 (0·69-1·79) for the same populations who were positive by tuberculin skin test. Pooled proportion of disease progression for individuals who were positive by IFN-γ release assay and tuberculin skin test was 6·1 (95% CI 2·3-11·5). Pooled RR for rates of disease progression in individuals who were positive by IFN-γ release assay and tuberculin skin test who were untreated versus those who were treated was 7·84 (95% CI 4·44-13·83). INTERPRETATION: IFN-γ release assays have a better predictive ability than tuberculin skin tests. Individuals who are positive by IFN-γ release assay might benefit from preventive treatment, but those who are positive by tuberculin skin test probably will not. Dual testing might improve detection, but further confirmation is needed. FUNDING: National Natural Science Foundation of China and Natural Foundation of Yunnan Province.


Subject(s)
Interferon-gamma/metabolism , Latent Tuberculosis/diagnosis , Tuberculin Test , Antitubercular Agents/therapeutic use , Humans , Latent Tuberculosis/drug therapy
4.
J Neuropathol Exp Neurol ; 79(5): 518-529, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32196082

ABSTRACT

Borrelia burgdorferi (Bb), which is neurotropic, can attack the central nervous system (CNS), leading to the development of various neurologic symptoms. The pathogenesis of Lyme neuroborreliosis (LNB) remains poorly understood. Presently, there is a lack of knowledge of the changes in mRNA and proteins in the CNS following early disseminated Lyme disease. Explants from the frontal cortex of 3 rhesus brains were incubated with medium alone or with medium containing live Bb for 6, 12, or 24 hours. Then, we analyzed identified mRNA and proteins in the frontal cortex tissues, allowing for an in-depth view of the transcriptome and proteome for a macroscopic and unbiased understanding of early disseminated Lyme disease in the brain. Through bioinformatics analysis, a complex network of enriched pathways that were mobilized during the progression of Lyme spirochete infection was described. Furthermore, based on the analysis of omics data, translational regulation, glycosaminoglycan/proteoglycan-binding activity in colonization and dissemination to tissues, disease-associated genes, and synaptic function were enriched, which potentially play a role in pathogenesis during the interaction between frontal cortex tissues and spirochetes. These integrated omics results provide unbiased and comprehensive information for the further understanding of the molecular mechanisms of LNB.


Subject(s)
Frontal Lobe/metabolism , Frontal Lobe/microbiology , Gene Expression Profiling , Lyme Disease/metabolism , Proteomics , Animals , Female , Gene Expression , Macaca mulatta , Male , RNA, Messenger/metabolism
5.
Curr Protein Pept Sci ; 21(5): 517-526, 2020.
Article in English | MEDLINE | ID: mdl-31613726

ABSTRACT

Lyme disease (LD) is an infectious disease caused by the spirochetes of genus borrelia, which are transmitted by the ticks of the genus ixodes. LD is transmitted by the spirochete B. burgdorferi sensu lato. Once in contact with the host through a tick bite, the pathogen comes into contact with the host defense, and must escape this machinery to establish LD, thus using a large number of mechanisms involving the vector of the pathogen, the pathogen itself and also the host. The initial diagnosis of the disease can be made based on the clinical symptoms of LD and the disease can be treated and cured with antibiotics if the diagnosis is made early in the beginning of the disease. Contrariwise, if LD is left untreated, the pathogen disseminates throughout the tissues and organs of the body, where it establishes different types of disease manifestations. In the nervous system, the inflammation caused by B. burgdorferi is known as Lyme neuroborreliosis (LNB). LNB is one of the principal manifestations of LD. In this review, we systematically describe the different molecular interactions among B. burgdorferi, the vector (tick) and the mammalian host.


Subject(s)
Arachnid Vectors/microbiology , Bacterial Proteins/genetics , Borrelia burgdorferi/pathogenicity , Host-Pathogen Interactions/genetics , Ixodes/microbiology , Lyme Disease/genetics , Membrane Proteins/genetics , Receptors, Cell Surface/genetics , Animals , Arachnid Vectors/immunology , Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Humans , Ixodes/immunology , Lyme Disease/immunology , Lyme Disease/microbiology , Lyme Disease/pathology , Membrane Proteins/immunology , Nervous System/immunology , Nervous System/microbiology , Nervous System/pathology , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Cell Surface/immunology , Saliva/microbiology , Signal Transduction
6.
Front Immunol ; 10: 2089, 2019.
Article in English | MEDLINE | ID: mdl-31552037

ABSTRACT

Background: Currently, there is no tuberculosis (TB) vaccine recommended for use in latent TB infections and healthy adults. M72/AS01E is a new peptide vaccine currently under development, which may improve protection against TB disease. This vaccine has been investigated in several phase I/II clinical trials. We conducted a meta-analysis to clarify the immunogenicity and safety of the M72/AS01E peptide vaccine. Methods: We searched the PubMed, Embase, and Cochrane Library databases for published studies (until December 2018) investigating this candidate vaccine. A meta-analysis was performed using the standard methods and procedures established by the Cochrane Collaboration. Results: Seven eligible studies-involving 4,590 participants-were selected. The analysis revealed a vaccine efficacy was 57.0%, significantly higher abundance of polyfunctional M72-specific CD4+ T cells [standardized mean difference (SMD) = 2.58] in the vaccine group vs. the control group, the highest seropositivity rate [relative risk (RR) = 74.87] at 1 month after the second dose of vaccination (Day 60), and sustained elevated anti-M72 IgG geometric mean concentration at study end (Day 210) (SWD = 4.94). Compared with the control, participants who received vaccination were at increased risk of local injection site redness [relative risk (RR) = 5.99], local swelling (RR = 7.57), malaise (RR = 3.01), and fatigue (RR = 3.17). However, they were not at increased risk of headache (RR = 1.57), myalgia (RR = 0.97), and pain (RR = 3.02). Conclusion: The M72/AS01E vaccine against TB is safe and effective. Although the vaccine is associated with a mild adverse reaction, it is promising for the prevention of TB in healthy adults.


Subject(s)
Immunogenicity, Vaccine , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Antibodies, Bacterial/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Immunoglobulin G/immunology , Outcome Assessment, Health Care , Randomized Controlled Trials as Topic , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/adverse effects
7.
Biomed Pharmacother ; 115: 108874, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31003080

ABSTRACT

Lyme disease, reffered to as Lyme borreliosis, is a tick-borne zoonotic disease caused by Borrelia burgdorferi spirochetes. Lyme arthritis, the most common, serious and harmful manifestation during the late stages of Lyme disease, is closely associated with the Borrelia burgdorferi basic membrane protein A (BmpA). Chemokines are also reported to have an important role in Lyme arthritis. Toll-like receptors (TLRs) recognize and bind to pathogen-associated molecules which are structurally conserved among microbes, to activate transcriptional events, including cytokine production, inflammation, and tissue damage. We speculated that BmpA could induce a storm of proinflammatory chemokines via TLRs and downstream moleculars, and that TLR1, TLR2, TLR5, TLR6 and the adaptor protein, MyD88, may be involved in this process. We explored this hypothesis using the human monocytic leukemia cell line, THP-1, and recombinant BmpA (rBmpA). Cell surface TLR1 and TLR2 were neutralized using specific antibodies before stimulation with rBmpA and analysis of chemokine secretion using a chemokine chip. Further, the expressions level of the four TLRs and MyD88 were analyzed following stimulation with rBmpA. Stimulation with rBmpA resulted in elevated levels of seven cytokines. Further, TLR1 and TLR2 antibody treated cells exhibited an overall reduction in rBmpA-induced chemokine expression. TLR1, TLR2, and MyD88 expression levels (both mRNA and protein) increased after stimulation with rBmpA. Our data confirm that TLR1, TLR2, and MyD88 are involved in BmpA-induced proinflammatory chemokines, which may be closely involved in Lyme arthritis pathogenesis.


Subject(s)
Bacterial Proteins/pharmacology , Borrelia burgdorferi/metabolism , Chemokines/metabolism , Macrophages/drug effects , Membrane Proteins/pharmacology , Toll-Like Receptor 2/metabolism , Chemokines/genetics , Gene Expression Regulation/drug effects , Humans , Macrophages/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Recombinant Proteins , THP-1 Cells , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics
8.
Front Immunol ; 10: 3067, 2019.
Article in English | MEDLINE | ID: mdl-31998324

ABSTRACT

Ixodes ticks are the main vectors for a number of zoonotic diseases, including Lyme disease. Ticks secrete saliva directly into a mammalian host while feeding on the host's blood. This action serves to modulate host immunity and coagulation, thus allowing ticks to attach and feed upon their host. One of the most extensively studied components of tick saliva is Salp15. Research has shown that this protein binds specifically to CD4 molecules on the surface of T lymphocytes, interferes with TCR-mediated signaling transduction, inhibits CD4+ T cell activation and proliferation, and impedes the secretion of interleukin 2 (IL-2). Salp15 also binds specifically to dendritic cell dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) to up-regulate the expression of CD73 in regulatory T cells. Collectively, these findings render this salivary protein a potential candidate for a range of therapeutic applications. Here, we discuss our current understanding of Salp15 and the mechanisms that might be used to treat disease.


Subject(s)
Saliva/immunology , Salivary Proteins and Peptides/immunology , Ticks/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Humans , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...