Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Language
Publication year range
1.
Emerg Infect Dis ; 30(5): 1055-1057, 2024 May.
Article in English | MEDLINE | ID: mdl-38666739

ABSTRACT

We report a clinical isolate of Burkholderia thailandensis 2022DZh obtained from a patient with an infected wound in southwest China. Genomic analysis indicates that this isolate clusters with B. thailandensis BPM, a human isolate from Chongqing, China. We recommend enhancing monitoring and surveillance for B. thailandensis infection in both humans and livestock.


Subject(s)
Burkholderia Infections , Burkholderia , Phylogeny , Wound Infection , Humans , Male , Burkholderia/genetics , Burkholderia/isolation & purification , Burkholderia/classification , Burkholderia Infections/microbiology , Burkholderia Infections/diagnosis , China/epidemiology , Genome, Bacterial , Wound Infection/microbiology , Middle Aged
3.
Front Microbiol ; 14: 1210319, 2023.
Article in English | MEDLINE | ID: mdl-37426023

ABSTRACT

Enterococcus faecalis is a well-established resident of the human gastrointestinal tract and is also a major cause of human infections. Unfortunately, therapeutic options for E. faecalis infections remain limited, particularly with the emergence of vancomycin-resistant strains in hospital settings. Consequently, there has been a renewed interest in phage therapy as an alternative to antibiotics. In this study, we have isolated a bacteriophage, vB_EfaS-SFQ1, from hospital sewage, which effectively infects E. faecalis strain EFS01. Phage SFQ1 is a siphovirus and exhibits a relatively broad host range. Furthermore, it has a short latent period of approximately 10 min and a large burst size of about 110 PFU/cell at a multiplicity of infection (MOI) of 0.01, and it could effectively disrupt the biofilms formed by E. faecalis. Thus, this study provides a detailed characterization of E. faecalis phage SFQ1, which has great potential for treating E. faecalis infections.

4.
Int J Med Sci ; 19(2): 402-415, 2022.
Article in English | MEDLINE | ID: mdl-35165525

ABSTRACT

Hypertension, diabetes mellitus, and coronary artery disease are common comorbidities and dangerous factors for infection and serious COVID-19. Polymorphisms in genes associated with comorbidities may help observe susceptibility and disease severity variation. However, specific genetic factors and the extent to which they can explain variation in susceptibility of severity are unclear. Therefore, we evaluated candidate genes associated with COVID-19 and hypertension, diabetes mellitus, and coronary artery disease. In particular, we performed searches against OMIM, NCBI, and other databases, protein-protein interaction network construction, and GO and KEGG pathway enrichment analyses. Results showed that the associated overlapping genes were TLR4, NLRP3, MBL2, IL6, IL1RN, IL1B, CX3CR1, CCR5, AGT, ACE, and F2. GO and KEGG analyses yielded 302 GO terms (q < 0.05) and 29 signaling pathways (q < 0.05), respectively, mainly including coronavirus disease-COVID-19 and cytokine-cytokine receptor interaction. IL6 and AGT were central in the PPI, with 8 and 5 connections, respectively. In this study, we identified 11 genes associated with both COVID-19 and three comorbidities that may contribute to infection and disease severity. The key genes IL6 and AGT are involved in regulating immune response, cytokine activity, and viral infection. Therefore, RAAS inhibitors, AGT antisense nucleotides, cytokine inhibitors, vitamin D, fenofibrate, and vaccines regulating non-immune and immune factors could be potential strategies to prevent and cure COVID-19. The study provides a basis for further investigation of genes and pathways with predictive value for the risk of infection and prognosis and could help guide drug and vaccine development to improve treatment efficacy and the development of personalised treatments, especially for COVID-19 individuals with common comorbidities.


Subject(s)
COVID-19/genetics , COVID-19/epidemiology , Comorbidity , Coronary Artery Disease/complications , Coronary Artery Disease/epidemiology , Coronary Artery Disease/genetics , Diabetes Complications/epidemiology , Diabetes Complications/genetics , Humans , Hypertension/complications , Hypertension/epidemiology , Hypertension/genetics , Mutation , Protein Interaction Maps
5.
Biol Res ; 51(1): 20, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29933757

ABSTRACT

Concerns have been raised about this article [1] relating to the appropriateness of the use of the shRNA (5'-GCGGAGGGTTTGAAAGAATATCTCGAGATATTCTTTCAAACCCTCCGCTTTTTT-3') as a non-targeting control and similarities in text and formatting with other published articles. This is currently under investigation and appropriate editorial action will be taken once the investigation is concluded. The authors did not respond to our correspondence regarding this expression of concern.

6.
Biol Res ; 49(1): 26, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27177590

ABSTRACT

BACKGROUND: Zinc finger RNA binding protein (ZFR) is involved in the regulation of growth and cancer development. However, little is known about ZFR function in pancreatic cancer. METHODS: Herein, to investigate whether ZFR is involved in tumor growth, Oncomine microarray data was firstly used to evaluate ZFR gene expression in human pancreatic tumors. Then short hairpin RNA (shRNA) targeting ZFR was designed and delivered into PANC-1 pancreatic cancer cells to knock down ZFR expression. Cell viability, cell proliferation and cell cycle analysis after ZFR knockdown were determined by MTT, colony forming and FACS, respectively. In addition, cell migration and invasion were assessed using the Transwell system. RESULTS: The expression of ZFR was significantly higher in pancreatic tumors than normal pancreas tissues by Oncomine database analysis. Knockdown of ZFR by shRNA-expressing lentivirus significantly decreased the viability and invasion ability of pancreatic cancer cells. Moreover, FACS analysis showed that knockdown of ZFR in PANC-1 cells caused a significant cell cycle arrest at G0/G1 phase. Furthermore, knockdown of ZFR decreased the levels of CDK2, CDK4, CyclinA and CyclinD1 and enhanced the expression of p27, which has evidenced by qRT-PCR and Western blot analysis. CONCLUSIONS: Knockdown of ZFR might provide a novel alternative to targeted therapy of pancreatic cancer and deserves further investigation.


Subject(s)
Gene Knockdown Techniques/methods , Pancreatic Neoplasms/pathology , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/metabolism , Animals , Blotting, Western , Cattle , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival , Cells, Cultured , Flow Cytometry/methods , Formazans , Humans , Lentivirus/genetics , Molecular Targeted Therapy , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA-Binding Proteins/genetics , Real-Time Polymerase Chain Reaction , Tetrazolium Salts
7.
Biol. Res ; 49: 1-8, 2016. ilus, graf
Article in English | LILACS | ID: biblio-950853

ABSTRACT

BACKGROUND: Zinc finger RNA binding protein (ZFR) is involved in the regulation of growth and cancer development. However, little is known about ZFR function in pancreatic cancer. METHODS: Herein, to investigate whether ZFR is involved in tumor growth, Oncomine microarray data was firstly used to evaluate ZFR gene expression in human pancreatic tumors. Then short hairpin RNA (shRNA) targeting ZFR was designed and delivered into PANC-1 pancreatic cancer cells to knock down ZFR expression. Cell viability, cell proliferation and cell cycle analysis after ZFR knockdown were determined by MTT, colony forming and FACS, respectively. In addition, cell migration and invasion were assessed using the Transwell system. RESULTS: The expression of ZFR was significantly higher in pancreatic tumors than normal pancreas tissues by Oncomine database analysis. Knockdown of ZFR by shRNA-expressing lentivirus significantly decreased the viability and invasion ability of pancreatic cancer cells. Moreover, FACS analysis showed that knockdown of ZFR in PANC-1 cells caused a significant cell cycle arrest at G0/G1 phase. Furthermore, knockdown of ZFR decreased the levels of CDK2, CDK4, CyclinA and CyclinD1 and enhanced the expression of p27, which has evidenced by qRT-PCR and Western blot analysis. CONCLUSIONS: Knockdown of ZFR might provide a novel alternative to targeted therapy of pancreatic cancer and deserves further investigation.


Subject(s)
Animals , Cattle , Humans , Pancreatic Neoplasms/pathology , RNA-Binding Proteins/metabolism , RNA, Small Interfering/pharmacology , Gene Knockdown Techniques/methods , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Tetrazolium Salts , Cell Survival , Cells, Cultured , Blotting, Western , RNA-Binding Proteins/genetics , Lentivirus/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Molecular Targeted Therapy , Real-Time Polymerase Chain Reaction , Flow Cytometry/methods , Formazans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...