Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cell Mol Biol Lett ; 29(1): 70, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741147

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis heat-resistant antigen (Mtb-HAg) is a peptide antigen released from the mycobacterial cytoplasm into the supernatant of Mycobacterium tuberculosis (Mtb) attenuated H37Ra strain after autoclaving at 121 °C for 20 min. Mtb-HAg can specifically induce γδ T-cell proliferation in vitro. However, the exact composition of Mtb-HAg and the protein antigens that are responsible for its function are currently unknown. METHODS: Mtb-HAg extracted from the Mtb H37Ra strain was subjected to LC‒MS mass spectrometry. Twelve of the identified protein fractions were recombinantly expressed in Escherichia coli by genetic engineering technology using pET-28a as a plasmid and purified by Ni-NTA agarose resin to stimulate peripheral blood mononuclear cells (PBMCs) from different healthy individuals. The proliferation of γδ T cells and major γδ T-cell subset types as well as the production of TNF-α and IFN-γ were determined by flow cytometry. Their proliferating γδ T cells were isolated and purified using MACS separation columns, and Mtb H37Ra-infected THP-1 was co-cultured with isolated and purified γδ T cells to quantify Mycobacterium viability by counting CFUs. RESULTS: In this study, Mtb-HAg from the attenuated Mtb H37Ra strain was analysed by LC‒MS mass spectrometry, and a total of 564 proteins were identified. Analysis of the identified protein fractions revealed that the major protein components included heat shock proteins and Mtb-specific antigenic proteins. Recombinant expression of 10 of these proteins in by Escherichia coli genetic engineering technology was used to successfully stimulate PBMCs from different healthy individuals, but 2 of the proteins, EsxJ and EsxA, were not expressed. Flow cytometry results showed that, compared with the IL-2 control, HspX, GroEL1, and GroES specifically induced γδ T-cell expansion, with Vγ2δ2 T cells as the main subset, and the secretion of the antimicrobial cytokines TNF-α and IFN-γ. In contrast, HtpG, DnaK, GroEL2, HbhA, Mpt63, EsxB, and EsxN were unable to promote γδ T-cell proliferation and the secretion of TNF-α and IFN-γ. None of the above recombinant proteins were able to induce the secretion of TNF-α and IFN-γ by αß T cells. In addition, TNF-α, IFN-γ-producing γδ T cells inhibit the growth of intracellular Mtb. CONCLUSION: Activated γδ T cells induced by Mtb-HAg components HspX, GroES, GroEL1 to produce TNF-α, IFN-γ modulate macrophages to inhibit intracellular Mtb growth. These data lay the foundation for subsequent studies on the mechanism by which Mtb-HAg induces γδ T-cell proliferation in vitro, as well as the development of preventive and therapeutic vaccines and rapid diagnostic reagents.


Subject(s)
Antigens, Bacterial , Cell Proliferation , Mycobacterium tuberculosis , T-Lymphocytes , Humans , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Antigens, Bacterial/genetics , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Tumor Necrosis Factor-alpha/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology
2.
Biochem Biophys Res Commun ; 718: 150083, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38735138

ABSTRACT

Acute lung injury (ALI) and its severe manifestation, acute respiratory distress syndrome (ARDS), represent critical clinical syndromes with multifactorial origins, notably stemming from sepsis within intensive care units (ICUs). Despite their high mortality rates, no selective cure is available beside ventilation support. Apoptosis plays a complex and pivotal role in the pathophysiology of acute lung injury. Excessive apoptosis of alveolar epithelial and microvascular endothelial cells can lead to disruption of lung epithelial barrier integrity, impairing the body's ability to exchange blood and gas. At the same time, apoptosis of damaged or dysfunctional cells, including endothelial and epithelial cells, can help maintain tissue integrity and accelerate recovery from organ pro-inflammatory stress. The balance between pro-survival and pro-apoptotic signals in lung injury determines patient outcomes, making the modulation of apoptosis an area of intense research in the quest for more effective therapies. Here we found that protein tyrosine phosphatase receptor type O (PTPRO), a poorly understood receptor-like protein tyrosine phosphatase, is consistently upregulated in multiple tissue types of mice under septic conditions and in the lung alveolar epithelial cells. PTPRO reduction by its selective short-interfering RNA (siRNA) leads to excessive apoptosis in lung alveolar epithelial cells without affecting cell proliferation. Consistently PTPRO overexpression by a DNA construct attenuates apoptotic signaling induced by LPS. These effects of PTPTO on cellular apoptosis are dependent on an ErbB2/PI3K/Akt/NFκB signaling pathway. Here we revealed a novel regulatory pathway of cellular apoptosis by PTPRO in lung alveolar epithelial cells during sepsis.


Subject(s)
Alveolar Epithelial Cells , Apoptosis , Lipopolysaccharides , Receptor-Like Protein Tyrosine Phosphatases, Class 3 , Animals , Humans , Male , Mice , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Apoptosis/drug effects , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Sepsis/metabolism , Sepsis/pathology , Signal Transduction/drug effects
3.
Cytokine ; 179: 156610, 2024 07.
Article in English | MEDLINE | ID: mdl-38640558

ABSTRACT

OBJECTIVES: To preliminarily assess the immunogenicity of Mtb-HAg in mice and the synergistic effect provided by HAg when co-immunised with BCG. METHODS: Mice were randomly grouped for different immunisations and then spleens were aseptically removed and lymphocytes were extracted for immediate detection of cytokines transcript levels and stimulation index(SI), cytokine secretion and multifunctional antigen-specific T cells were detected after incubation for different times. RESULTS: HAg extracted from active Mtb is a group of mixed polypeptides with molecular weights of (10-14) kDa. It can significantly stimulate lymphocytes proliferation and increase SI. Injection of HAg alone and in combination with BCG induced significantly higher numbers of multifunctional antigen-specific T cells including CD4+ IFN-γ+, CD4+ IL-2+, CD8+ IFN-γ+, and CD8+ IL-2+ cells than that in BCG-treated mice. Co-immunisation induced the secretion of higher levels of IFN-γ, TNF-α, IL-2 and IL-4 and increased their mRNA expression levels. Significant increases in the transcription levels of IL-10, IL-12 and IL-17 were observed in the co-immunised group with the assistance of HAg. CONCLUSION: We demonstrated that HAg has favourable immunogenicity, triggers a stronger Th1-type immune response and proposed the hypothesis that HAg can be used as a BCG booster to further enhance the benefits of BCG.


Subject(s)
Antigens, Bacterial , Cytokines , Mycobacterium tuberculosis , Animals , Mice , Antigens, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Cytokines/metabolism , Mycobacterium tuberculosis/immunology , Mycobacterium bovis/immunology , BCG Vaccine/immunology , Female , Mice, Inbred BALB C , Immunization/methods , Cell Proliferation/drug effects , Spleen/immunology
4.
J Microbiol ; 62(1): 49-62, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38337112

ABSTRACT

Tuberculosis (TB), a bacterial infectious disease caused by Mycobacterium tuberculosis (M. tuberculosis), is a significant global public health problem. Mycobacterium tuberculosis expresses a unique family of PE_PGRS proteins that have been implicated in pathogenesis. Despite numerous studies, the functions of most PE_PGRS proteins in the pathogenesis of mycobacterium infections remain unclear. PE_PGRS45 (Rv2615c) is only found in pathogenic mycobacteria. In this study, we successfully constructed a recombinant Mycobacterium smegmatis (M. smegmatis) strain which heterologously expresses the PE_PGRS45 protein. We found that overexpression of this cell wall-associated protein enhanced bacterial viability under stress in vitro and cell survival in macrophages. MS_PE_PGRS45 decreased the secretion of pro-inflammatory cytokines such as IL-1ß, IL-6, IL-12p40, and TNF-α. We also found that MS_PE_PGRS45 increased the expression of the anti-inflammatory cytokine IL-10 and altered macrophage-mediated immune responses. Furthermore, PE_PGRS45 enhanced the survival rate of M. smegmatis in macrophages by inhibiting cell apoptosis. Collectively, our findings show that PE_PGRS45 is a virulent factor actively involved in the interaction with the host macrophage.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Mycobacterium tuberculosis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Immunity, Innate , Cytokines/metabolism , Apoptosis , Mycobacterium smegmatis/genetics
5.
Front Cell Infect Microbiol ; 13: 1255905, 2023.
Article in English | MEDLINE | ID: mdl-37818041

ABSTRACT

Background: Mycobacterium tuberculosis antigen (Mtb-Ag) is a polypeptide component with a molecular weight of 10-14 kDa that is obtained from the supernatant of the H37Ra strain after heat treatment. It stimulates the activation and proliferation of γδT cells in the blood to produce an immune response against tuberculosis. Mtb-Ag is therefore crucial for classifying and detecting the central genes and key pathways involved in TB initiation and progression. Methods: In this study, we performed high-throughput RNA sequencing of peripheral blood mononuclear cells (PBMC) from Mtb-Ag-stimulated and control samples to identify differentially expressed genes and used them for gene ontology (GO) and a Kyoto Encyclopedia of Genomes (KEGG) enrichment analysis. Meanwhile, we used PPI protein interaction network and Cytoscape analysis to identify key genes and qRT-PCR to verify differential gene expression. Single-gene enrichment analysis (GSEA) was used further to elucidate the potential biological functions of key genes. Analysis of immune cell infiltration and correlation of key genes with immune cells after Mtb-Ag-stimulated using R language. Results: We identified 597 differentially expressed genes in Mtb-Ag stimulated PBMCs. KEGG and GSEA enrichment analyzed the cellular pathways related to immune function, and DEGs were found to be primarily involved in the TNF signaling pathway, the IL-17 signaling pathway, the JAK-STAT signaling pathway, cytokine-cytokine receptor interactions, and the NF-κB signaling pathway. Wayne analysis using GSEA, KEGG, and the protein-protein interaction (PPI) network showed that 34 genes, including PTGS2, IL-1ß, IL-6, TNF and IFN-γ et al., were co-expressed in the five pathways and all were up-regulated by Mtb-Ag stimulation. Twenty-four DEGs were identified using qRT-PCR, including fourteen up-regulated genes (SERPINB7, IL20, IFNG, CSF2, PTGS2, TNF-α, IL36G, IL6, IL10, IL1A, CXCL1, CXCL8, IL4, and CXCL3) and ten down-regulated genes (RTN1, CSF1R CD14, C5AR1, CXCL16, PLXNB2, OLIG1, EEPD1, ENG, and CCR1). These findings were consistent with the RNA-Seq results. Conclusion: The transcriptomic features associated with Mtb-Ag provide the scientific basis for exploring the intracellular immune mechanisms against Mtb. However, more studies on these DEGs in pathways associated with Mtb-Ag stimulation are needed to elucidate the underlying pathologic mechanisms of Mtb-Ag during Mtb infection.


Subject(s)
Mycobacterium tuberculosis , Serpins , Tuberculosis , Humans , Leukocytes, Mononuclear , Cyclooxygenase 2 , Cytokines/metabolism , Gene Expression Profiling/methods , Computational Biology/methods
6.
Front Immunol ; 14: 1255920, 2023.
Article in English | MEDLINE | ID: mdl-37841250

ABSTRACT

Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (Mtb), and its incidence and mortality are increasing. The BCG vaccine was developed in the early 20th century. As the most widely administered vaccine in the world, approximately 100 million newborns are vaccinated with BCG every year, which has saved tens of millions of lives. However, due to differences in region and race, the average protective rate of BCG in preventing tuberculosis in children is still not high in some areas. Moreover, because the immune memory induced by BCG will weaken with the increase of age, it is slightly inferior in preventing adult tuberculosis, and BCG revaccination cannot reduce the incidence of tuberculosis again. Research on the mechanism of Mtb and the development of new vaccines against TB are the main strategies for preventing and treating TB. In recent years, Pro-Glu motif-containing (PE) and Pro-Pro-Glu motif-containing (PPE) family proteins have been found to have an increasingly important role in the pathogenesis and chronic protracted infection observed in TB. The development and clinical trials of vaccines based on Mtb antigens are in progress. Herein, we review the immunological effects of PE/PPE proteins and the development of common PE/PPE vaccines.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis Vaccines , Tuberculosis , Infant, Newborn , Child , Humans , BCG Vaccine , Tuberculosis/prevention & control , Bacterial Proteins
7.
Am J Transl Res ; 15(8): 5159-5167, 2023.
Article in English | MEDLINE | ID: mdl-37692935

ABSTRACT

OBJECTS: Epidemiologic studies have linked exposure to airborne pollutant particulate matter (PM) with increased rates of chronic cardiopulmonary diseases, including asthma and idiopathic pulmonary fibrosis (IPF). Several investigations have suggested that the epithelial-to-mesenchymal transition (EMT) may contribute to the complex pathobiology of environmental exposure-mediated pulmonary fibrosis. The present study was designed to characterize the mechanisms of PM-mediated EMT in human lung epithelial cells (HBECs). METHODS AND RESULTS: PM induced significant dose (0-100 µg/ml) and time (0-72 h)-dependent increases in transforming growth factor ß (TGFß) and fibronectin (FN) protein levels in HBECs lysates. PM-activated TGFß and FN protein production in HBECs was prevented by the antioxidant N-acetyl-cysteine (NAC, 5 mM). Furthermore, the NF-κB inhibitor BAY11-7082 (5 µM) abolished PM-induced FN production in HBECs. Biomarkers of EMT (ACTA2, SNAIL1 and SNAIL2) in PM-treated HBECs were significantly increased at the mRNA level compared to control cells. CONCLUSIONS: These results demonstrate that PM increases protein levels of TGFß and FN via reactive oxygen species (ROS)-dependent pathways. In addition, PM exposure induces EMT in human lung epithelial cells, supporting a novel mechanism for PM-induced pulmonary fibrosis.

8.
DNA Cell Biol ; 42(5): 254-264, 2023 May.
Article in English | MEDLINE | ID: mdl-37074124

ABSTRACT

Tuberculosis is an important chronic and often fatal infectious disease mainly caused by the bacterium Mycobacterium tuberculosis (Mtb). Mtb is one of the most successful pathogens that harbors several potential virulence factors not found in nonpathogenic mycobacteria. As the Mtb cell envelope is closely associated with its virulence and resistance, it is very important to understand the cell envelope for better treatment of causative pathogen. There is increasing evidence that Pro-Glu (PE) and Pro-Pro-Glu (PPE) proteins are the major effectors of virulence and persistence encoded in the Mtb H37Rv genome. However, the function of PE8 has not been explored to date. In this study, we heterologously expressed PE8 in nonpathogenic, fast-growing M. smegmatis to investigate the interaction between PE8 and the host to determine its possible biological functions. We found that recombinant M. smegmatis cells expressing PE8 were less susceptible to sodium dodecyl sulfate-induced surface stress compared with those expressing the empty vector, suggesting that PE8 may be involved in stress responses. In addition, macrophages infected with PE8-expressing M. smegmatis produced obviously lower levels of the proinflammatory factor IL-1ß, IL-6, and TNF-α and higher levels of the inhibitory factor IL-10. We further found that PE8 promoted M. smegmatis survival within macrophages by inhibiting late apoptosis of macrophages. Collectively, selective targeting of the PE/PPE protein family offers an untapped opportunity to the development of more effective and safer drugs against Mtb infection.


Subject(s)
Cytokines , Mycobacterium tuberculosis , Cytokines/genetics , Cytokines/metabolism , Mycobacterium tuberculosis/metabolism , Bacterial Proteins/genetics , Host-Pathogen Interactions , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Apoptosis
9.
Biol Pharm Bull ; 46(1): 42-51, 2023.
Article in English | MEDLINE | ID: mdl-36596525

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide, synthesizing and screening of novel anti-cancer drugs provides an alternative therapeutic strategy for renewal of the chemotherapy regimens against lung cancer. To this end, several compounds were synthesized based on the modification of the original myricetin, and their anti-tumor activity against the human non-small cell lung cancer (NSCLC) A549 cells were measured. Among the myricetin derivatives, S4-10 has displayed the highest antitumor efficacy in dose-dependent manner. The proliferation of A549 cells were significantly attenuated by given 6 µM of S4-10 both in vitro and in vivo. Further, the treatment of S4-10 also results in the inhibition of cell migration and invasiveness and the induction of cell apoptosis and G2 cycle arrest of A549 cells. Moreover, we found that S4-10 inhibits the progression of A549 cells through the sterol biosynthetic-cell apoptosis axis. These findings shed the light of developing S4-10 as a promising treatment agent for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , A549 Cells , Cell Cycle Checkpoints , Apoptosis , Cell Proliferation , Cell Line, Tumor , Cell Cycle
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(12): 1091-1096, 2022 Dec.
Article in Chinese | MEDLINE | ID: mdl-36585231

ABSTRACT

Objective To investigate the effect of protein tyrosine phosphatase receptor type O (PTPRO) on the phagocytic activity of alveolar epithelial cells in LPS-induced acute lung injury. Methods Mice were randomly divided into the normal control group and LPS stimulation group. The infiltration of inflammatory cells was detected by HE staining. The cytokine TNF-α level in lung was analyzed by ELISA. Western blotting was performed to detect the effect of LPS on PTPRO protein expression in lung. After the expression of PTPRO in MLE-12 cells was silenced by siRNA in vitro, flow cytometry was used to detect the effects of LPS and PTPRO siRNA on the phagocytic activity of MLE-12 cells, and the effects of LPS and PTPRO siRNA on the expression of PTPRO, AKT and phosphorylated AKT protein were measured by Western blotting. Results After the establishment of murine acute lung injury model by LPS injection(1 mg/kg), the infiltrated polymorphonuclear leukocytes were markedly increased. The level of TNF-α in lung tissue and the expression of PTPRO in MLE-12 cells were both significantly increased after LPS stimulation. However, the activity of MLE-12 cells to phagocytose fluorescent microbeads was evidently decreased after silencing PTPRO. Furthermore, silencing PTPRO induced a remarkable decrease in the phosphorylation of AKT in MLE-12 cells. Conclusion PTPRO can promote phagocytic activity of MLE-12 cells via activating AKT signaling pathway.


Subject(s)
Acute Lung Injury , Alveolar Epithelial Cells , Mice , Animals , Alveolar Epithelial Cells/metabolism , Lipopolysaccharides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Signal Transduction , Protein Tyrosine Phosphatases/adverse effects , Protein Tyrosine Phosphatases/metabolism
11.
Free Radic Biol Med ; 193(Pt 1): 177-189, 2022 11 20.
Article in English | MEDLINE | ID: mdl-36244589

ABSTRACT

Tuberculosis (TB), caused by mycobacterium tuberculosis (M. tuberculosis) infection, is one of the leading causes of death globally and poses a threat to public health. During infection, M. tuberculosis causes redox imbalance and dysfunctions of protective immunity. Transcription factor nuclear factor erythroid 2 (NF-E2)-related factor (Nrf2) is a major modulator of cellular redox homeostasis via transcriptional induction of cytoprotective genes to protect cell against the damage from insults. Thus, we hypothesize that Nrf2 may regulate protective immunity against M. tuberculosis. RNA-seq and immunoblotting results suggested that the expression of Nrf2 protein increased after M. tuberculosis infection, and decreased upon long-term M. tuberculosis infection, while Keap1 protein maintained a low expression level during M. tuberculosis infection. Furthermore, Nrf2 activator sulforaphane (SFN) decreased proinflammatory cytokines production, phagocytosis and host cell apoptosis, while increasing ROS levels and promoting autophagy in THP1 macrophages infected with M. tuberculosis. In addition, SFN-activated Nrf2 augmented bacterial killing by macrophages, which might be due to the regulation of protective immunity via Nrf2. Combined, our results extend the understanding of the complex innate immunity regulation by Nrf2 against mycobacterial infection. Also, these findings suggested that the regulation of Nrf2 signaling cascade could be used as a therapeutic target for the treatment of TB patients and the development of better anti-TB vaccines.


Subject(s)
Macrophages , Mycobacterium tuberculosis , NF-E2-Related Factor 2 , Tuberculosis , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Macrophages/metabolism , Macrophages/microbiology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Tuberculosis/genetics , Tuberculosis/metabolism , THP-1 Cells
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(9): 789-793, 2022 Sep.
Article in Chinese | MEDLINE | ID: mdl-36082708

ABSTRACT

Objective To establish a THP-1 macrophage model infected by Mycobacterium smegmatis expressing green fluorescent protein (GFP), to quickly locate and visually detect Mycobacterium smegmatis, and to provide a tracer tool to identify the pathogenesis of tuberculosis and develop new tuberculosis vaccines. Methods The enhanced green fluorescent protein (EGFP) gene sequence was amplified by PCR using pEGFP-N1 plasmid as a template to obtain the coding gene of EGFP, and the amplified product was cloned into the vector pALACE to establish the recombinant plasmid pALACE-EGFP. Electroporation transformed the pALACE-EGFP into Mycobacterium smegmatis, and recombinant Mycobacterium smegmatis clones were screened by hygromycin resistance. After expanded culture, the smears were observed by fluorescence microscopy. The THP-1 macrophages were infected with recombinant Mycobacterium smegmatis, and the expression of EGFP was observed. Results The recombinant plasmid pALACE-EGFP was constructed appropriately. The recombinant Mycobacterium smegmatis was observed under fluorescence microscope. And it was confirmed that EGFP was expressed in recombinant Mycobacterium smegmatis, and THP-1 macrophages emitted green fluorescence after infection. Conclusion The successful establishment of recombinant Mycobacterium smegmatis expressing EGFP protein provides insights for investigating infection and pathogenesis of Mycobacterium tuberculosis.


Subject(s)
Mycobacterium smegmatis , Mycobacterium tuberculosis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Macrophages/metabolism , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Plasmids/genetics
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(1): 78-83, 2022 Jan.
Article in Chinese | MEDLINE | ID: mdl-35078578

ABSTRACT

Objective To clone, express and purify PPE15 recombinant protein from Mycobacterium tuberculosis (H37Rv), as well as prepare and characterize its rabbit polyclonal antibody. Methods By The PPE15 gene was amplified from the genome of Mycobacterium tuberculosis H37Rv by PCR, and the His-tagged prokaryotic PPE15 prokaryotic expression plasmid pET28a-PPE15 was constructed by homologous recombination cloning technique, and transformed into E. coli BL21 (DE3). PPE15 expression was induced by isopropyl-ß-D-thiogalactopyranoside (IPTG). Recombinant PPE15 was identified by SDS-PAGE, and further purified by affinity chromatography with a Ni-NTA column. The renaturation purified PPE15 protein was used to immunize New-Zealand rabbit to prepare polyclonal antibodies. The antibody specificity was analyzed by Western blot analysis, and antibody titer was determined by indirect ELISA. Results Recombinant prokaryotic PPE15 protein was successfully expressed and purified with a molecular weight of 38 kDa. The purified PPE15 protein exhibited positive reaction with the serum of TB patients and the PPE15 protein, the titer of the polyclonal antibodies reaches more than 1:1 300 480. Conclusion The recombinant protein PPE15 was successfully expressed and purified, and high titer rabbit-derived polyclonal antibody was prepared which provided an experimental basis for further functional studies of PPE15 protein.


Subject(s)
Mycobacterium tuberculosis , Animals , Antibodies , Antibody Specificity , Blotting, Western , Escherichia coli/genetics , Humans , Mycobacterium tuberculosis/genetics , Rabbits
14.
J Microbiol Biotechnol ; 31(11): 1583-1590, 2021 Nov 28.
Article in English | MEDLINE | ID: mdl-34489370

ABSTRACT

Studies have demonstrated that PE_PGRS45 is constitutively expressed under various environmental conditions (such as nutrient depletion, hypoxia, and low pH) of the in vitro growth conditions examined, indicating that PE_PGRS45 protein is critical to the basic functions of Mycobacterium tuberculosis. However, there are few reports about the biochemical function and pathogenic mechanism of PE_PGRS45 protein. The fact that this M. tuberculosis gene is not easily expressed in E. coli may be mainly due to the high content of G+C and the use of unique codons. Fusion tags are indispensable tools used to improve the soluble expression of recombinant proteins and accelerate the characterization of protein structure and function. In the present study, His6, Trx, and His6-MBP were used as fusion tags, but only MBP-PE_PGRS45 was expressed solubly. The purification using His6-MBP tag-specific binding to the Ni column was easy to separate after the tag cleavage. We used the purified PE_PGRS45 to immunize New Zealand rabbits and obtained anti- PE_PGRS45 serum. We found that the titer of polyclonal antibodies against PE_PGR45 was higher than 1:256000. The result shows that purified PE_PGRS45 can induce New Zealand rabbits to produce high-titer antibodies. In conclusion, the recombinant protein PE_PGRS45 was successfully expressed in E. coli and specific antiserum was prepared, which will be followed by further evaluation of these specific antigens to develop highly sensitive and specific diagnostic tests for tuberculosis.


Subject(s)
Antibodies/metabolism , Codon , Genes, Bacterial , Mycobacterium tuberculosis/genetics , Animals , Antibody Formation , Base Sequence , Escherichia coli , Immune Sera/biosynthesis , Rabbits , Recombinant Proteins/biosynthesis , Sequence Alignment
15.
Front Oncol ; 11: 691762, 2021.
Article in English | MEDLINE | ID: mdl-34367973

ABSTRACT

BACKGROUND: Tumor cells initiate hypoxia-induced mechanisms to fuel cell proliferation, invasion, and metastasis, largely mediated by low O2-responsive Hypoxia-Inducible Factor 1 Alpha (HIF-1α). Therefore, hyperbaric oxygen therapy (HBO) is now being studied in cancer patients, but its impact upon non-small-cell lung cancer (NSCLC) cell metabolism remains uncharacterized. METHODS: We employed the NSCLC cell lines A549 and H1299 for in vitro studies. Glucose uptake, pyruvate, lactate, and adenosine triphosphate (ATP) assays were used to assess aerobic glycolysis (Warburg effect). A quantitative glycolytic flux model was used to analyze the flux contributions of HIF-1α-induced glucose metabolism genes. We used a Lewis lung carcinoma (LLC) murine model to measure lung tumorigenesis in C57BL/6J mice. RESULTS: HBO suppressed hypoxia-induced HIF-1α expression and downstream HIF-1α signaling in NSCLC cells. One HIF-1α-induced glucose metabolism gene-Phosphofructokinase, Platelet (PFKP)-most profoundly enhanced glycolytic flux under both low- and high-glucose conditions. HBO suppressed hypoxia-induced PFKP transactivation and gene expression via HIF-1α downregulation. HBO's suppression of the Warburg effect, suppression of hyperproliferation, and suppression of epithelial-to-mesenchymal transition (EMT) in hypoxic NSCLC cell lines is mediated by the HIF-1α/PFKP axis. In vivo, HBO therapy inhibited murine LLC lung tumor growth in a Pfkp-dependent manner. CONCLUSIONS: HBO's repression of the Warburg effect, repression of hyperproliferation, and repression of EMT in hypoxic NSCLC cells is dependent upon HIF-1α downregulation. HIF-1α's target gene PFKP functions as a central mediator of HBO's effects in hypoxic NSCLC cells and may represent a metabolic vulnerability in NSCLC tumors.

16.
Emerg Microbes Infect ; 10(1): 578-588, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33666534

ABSTRACT

Mycobacterium tuberculosis (M. tuberculosis) is the pathogen which causes tuberculosis (TB), a significant human public health threat. Co-infection of M. tuberculosis and the human immunodeficiency virus (HIV), emergence of drug resistant M. tuberculosis, and failure to develop highly effective TB vaccines have limited control of the TB epidemic. Trained immunity is an enhanced innate immune response which functions independently of the adaptive/acquired immune system and responds non-specifically to reinfection with invading agents. Recently, several studies have found trained immunity has the capability to control and eliminate M. tuberculosis infection. Over the past decades, however, the consensus was adaptive immunity is the only protective mechanism by which hosts inhibit M. tuberculosis growth. Furthermore, autophagy plays an essential role in the development of trained immunity. Further investigation of trained immunity, M. tuberculosis infection, and the role of autophagy in this process provide new possibilities for vaccine development. In this review, we present the general characteristics of trained immunity and autophagy. We additionally summarize several examples where initiation of trained immunity contributes to the prevention of M. tuberculosis infection and propose future directions for research in this area.


Subject(s)
Autophagy , Immunity, Innate , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Adaptive Immunity , Animals , Humans , Immunologic Memory , Vaccination
17.
Mol Cancer ; 19(1): 159, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33176804

ABSTRACT

One unmet challenge in lung cancer diagnosis is to accurately differentiate lung cancer from other lung diseases with similar clinical symptoms and radiological features, such as pulmonary tuberculosis (TB). To identify reliable biomarkers for lung cancer screening, we leverage the recently discovered non-canonical small non-coding RNAs (i.e., tRNA-derived small RNAs [tsRNAs], rRNA-derived small RNAs [rsRNAs], and YRNA-derived small RNAs [ysRNAs]) in human peripheral blood mononuclear cells and develop a molecular signature composed of distinct ts/rs/ysRNAs (TRY-RNA). Our TRY-RNA signature precisely discriminates between control, lung cancer, and pulmonary TB subjects in both the discovery and validation cohorts and outperforms microRNA-based biomarkers, which bears the diagnostic potential for lung cancer screening.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Leukocytes, Mononuclear/metabolism , Lung Neoplasms/diagnosis , RNA, Small Untranslated/genetics , Case-Control Studies , Cohort Studies , Humans , Lung Neoplasms/blood , Lung Neoplasms/genetics , Prognosis , RNA, Small Untranslated/blood
18.
Front Immunol ; 11: 1585, 2020.
Article in English | MEDLINE | ID: mdl-32793225

ABSTRACT

To maintain alveolar gas exchange, the alveolar surface has to limit unnecessary inflammatory responses. This involves crosstalk between alveolar epithelial cells (AECs) and alveolar macrophages (AMs) in response to damaging factors. We recently showed that insulin-like growth factor (IGF)-1 regulates the phagocytosis of AECs. AMs secrete IGF-1 into the bronchoalveolar lavage fluid (BALF) in response to inflammatory stimuli. However, whether AECs regulate the production of IGF-1 by AMs in response to inflammatory signals remains unclear, as well as the role of IGF-1 in controlling the alveolar balance in the crosstalk between AMs and AECs under inflammatory conditions. In this study, we demonstrated that IGF-1 was upregulated in BALF and lung tissues of acute lung injury (ALI) mice, and that the increased IGF-1 was mainly derived from AMs. In vitro experiments showed that the production and secretion of IGF-1 by AMs as well as the expression of TGF-ß were increased in LPS-stimulated AEC-conditioned medium (AEC-CM). Pharmacological blocking of TGF-ß in AECs and addition of TGF-ß neutralizing antibody to AEC-CM suggested that this AEC-derived cytokine mediates the increased production and secretion of IGF-1 from AMs. Blocking TGF-ß synthesis or treatment with TGF-ß neutralizing antibody attenuated the increase of IGF-1 in BALF in ALI mice. TGF-ß induced the production of IGF-1 by AMs through the PI3K/Akt signaling pathway. IGF-1 prevented LPS-induced p38 MAPK activation and the expression of the inflammatory factors MCP-1, TNF-α, and IL-1ß in AECs. However, IGF-1 upregulated PPARγ to increase the phagocytosis of apoptotic cells by AECs. Intratracheal instillation of IGF-1 decreased the number of polymorphonuclear neutrophils in BALF of ALI model mice, reduced alveolar congestion and edema, and suppressed inflammatory cell infiltration in lung tissues. These results elucidated a mechanism by which AECs used TGF-ß to regulate IGF-1 production from AMs to attenuate endogenous inflammatory signals during alveolar inflammation.


Subject(s)
Alveolar Epithelial Cells/metabolism , Insulin-Like Growth Factor I/biosynthesis , Macrophages, Alveolar/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/immunology , Cell Communication , Disease Models, Animal , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Mice , Phagocytosis/immunology , Phosphatidylinositol 3-Kinases/metabolism , Pneumonia/etiology , Pneumonia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 36(6): 549-554, 2020 Jun.
Article in Chinese | MEDLINE | ID: mdl-32696746

ABSTRACT

Objective To clone proline-glutamate 8 (PE8) gene segment from Mycobacterium tuberculosis (H37Rv), construct the recombinant plasmid pET28a-PE8, express recombinant PE8 protein, and prepare its polyclonal antibody. Methods Using a standard homologous recombination cloning technology, we cloned the PE8 gene into the prokaryotic vector pET28a. After sequence confirmation, it was transformed into E. coli BL21 (DE3) and treated with 0.5 mmol/L isopropyl-beta-D-thiogalactopyranoside (IPTG) to induce protein expression. We purified and renatured the recombinant PE8 protein, and immunized New Zealand rabbits to prepare the polyclonal antibody. Antibody titer was determined by indirect ELISA and the specificity was evaluated by Western blot analysis. Results The recombinant plasmid pET28a-PE8 was successfully constructed, and the PE8 protein was primarily expressed in an inclusion body in E. coli. After renaturation and purification, a purity of about 90% of the recombinant protein was achieved. The titer of the polyclonal antibody was higher than 1:430 080. The polyclonal antibody could specifically recognize the recombinant PE8 protein. Conclusion We have successfully expressed and purified recombinant PE8 protein, which can be further utilized to generate PE8 polyclonal antibody with acceptable titer and specificity.


Subject(s)
Mycobacterium tuberculosis , Animals , Antibodies , Antibody Specificity , Bacterial Proteins , Blotting, Western , Escherichia coli/genetics , Mycobacterium tuberculosis/genetics , Rabbits
20.
Biochem Biophys Res Commun ; 529(3): 839-845, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32616311

ABSTRACT

Sepsis-associated encephalopathy (SAE) is a devastating neurological complication of sepsis with intolerable high motility. SAE is accompanied with brain vascular injury, endothelial hyperpermeability, and neutrophil infiltration into the brain tissue, key inflammatory processes leading to further brain edema and neuronal cell apoptosis. Recent studies from us and others suggest that the chemokine receptor C-X-C Motif Chemokine Receptor 2 (CXCR2) is crucial for neutrophil recruitment during SAE. Here we use CXCR2 antagonist SB225002 to characterize the role of CXCR2 in brain infiltration of neutrophil in a murine model of SAE. Systemic administration of high-dose LPS (10 mg/kg) induced evident neutrophil infiltration into the cerebral cortex in wild-type mice. However, CXCR2 antagonist SB225002 markedly attenuated neutrophil infiltration into brain. The CXCR2 expression on neutrophils in the peripheral circulation was dramatically downregulated in response to this LPS dose, and endothelial CXCR2 was significantly upregulated, suggesting endothelial but not neutrophil CXCR2 plays a more important role in neutrophil infiltration into brain. Strikingly, although these CXCR2 antagonist SB225002 treated mice displayed reduced neutrophil infiltration, no change in neutrophil rolling and adhesion was observed. Furthermore, we confirmed that CXCR2 agonist CXCL1 induced a marked increase in actin stress fiber synthesis and paracellular gap formation in cultured cerebral endothelial cells, which is attenuated by SB225002. Thus, these results demonstrate a selective role for endothelial CXCR2 to regulate cerebral vascular permeability and neutrophil transmigration in high-dose LPS induced neuroinflammation, and also suggest a therapeutic potential of CXCR2 antagonist SB225002 in SAE.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Phenylurea Compounds/therapeutic use , Receptors, Interleukin-8B/antagonists & inhibitors , Sepsis-Associated Encephalopathy/drug therapy , Animals , Brain/drug effects , Brain/immunology , Brain Edema , Cell Line , Disease Models, Animal , Lipopolysaccharides/immunology , Male , Mice, Inbred C57BL , Neutrophils/immunology , Receptors, Interleukin-8B/immunology , Sepsis-Associated Encephalopathy/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...