Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Zhongguo Fei Ai Za Zhi ; 27(5): 337-344, 2024 May 20.
Article in Chinese | MEDLINE | ID: mdl-38880921

ABSTRACT

BACKGROUND: Lung adenocarcinoma (LUAD) is a highly morbid and fatal cancer. Despite advancements in modern medical treatment, the 5-year survival rate of patients remains suboptimal. Our previous study revealed that zinc finger SWIM-type containing 1 (ZSWIM1), a novel protein, promotes the proliferation, migration, and invasion of LUAD cells. The aim of this study is to investigate the impact of E3 ubiquitin ligase tripartite motif protein 21 (TRIM21) on ZSWIM1-mediated cell proliferation and migration. METHODS: The interaction and co-localization between TRIM21 and ZSWIM1 were verified using co-immunoprecipitation (Co-IP) and immunofluorescence (IF). The effects of TRIM21 and ZSWIM1 on the proliferation and migration of LUAD cells were assessed through MTT and Transwell assays, respectively. Western blot (WB) analysis was conducted to evaluate the impact of TRIM21 and ZSWIM1 on the expression of epithelial-mesenchymal transition (EMT) markers in LUAD cells. The influence of TRIM21 on the ubiquitination of ZSWIM1 was examined using Co-IP combined with WB. RESULTS: TRIM21 was found to interact and co-localize with ZSWIM1. Overexpression of TRIM21 inhibited the proliferation and migration of LUAD cells. Overexpression of TRIM21 reduced the promoting effect of ZSWIM1 on the proliferation, migration, and invasion of lung adenocarcinoma cells, and reversed the impact of ZSWIM1 on the expression of E-cadherin and Vimentin. Conversely, knockdown of TRIM21 further enhanced the promoting effect of ZSWIM1 on the proliferation and migration of LUAD cells. Mechanistically, we observed that overexpression of TRIM21 significantly enhanced the ubiquitination level of ZSWIM1, leading to a decrease in ZSWIM1 protein expression. CONCLUSIONS: TRIM21 binds to and promotes the ubiquitination of ZSWIM1, resulting in reduced protein expression of ZSWIM1, which leads to the inhibition of ZSWIM1-mediated promotion of proliferation, migration, and invasion in LUAD cells.


Subject(s)
Adenocarcinoma of Lung , Cell Movement , Cell Proliferation , Lung Neoplasms , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Epithelial-Mesenchymal Transition , Cell Line, Tumor , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Ubiquitination , Protein Binding , A549 Cells
2.
Immunology ; 170(2): 230-242, 2023 10.
Article in English | MEDLINE | ID: mdl-37259771

ABSTRACT

Antibody inhibitors that block PD-1/PD-L1 interaction have been approved for oncological clinics, yielding impressive treatment effects. Small molecules inhibiting PD-1 signalling are at various stages of development, given that small molecular drugs are expected to outperform protein drugs in several ways. Currently, a significant portion of these small molecular inhibitors achieve this purpose by binding to a limited region of the PD-L1 protein, thereby limiting the choice of chemical structures. Alternative strategies for developing small-molecular PD-1 inhibitors are urgently needed to broaden the choice of chemical structures. Here, we report that 6-mercaptopurine (6-MP) inhibits PD-1 signalling, activates T cell function in vitro and in vivo and shrinks tumours by activating cytotoxic T cells. Mechanistically, 6-MP potently inhibited PD-1 signalling by blocking the recruitment of SHP2 by PD-1. Considering that 6-MP is a chemotherapeutic agent already approved by the FDA for childhood leukaemia, our work revealed a novel anti-tumour mechanism for this drug and suggests that 6-MP warrants further clinical evaluation for other tumour types.


Subject(s)
Mercaptopurine , Neoplasms , Humans , Child , Mercaptopurine/pharmacology , Mercaptopurine/therapeutic use , Programmed Cell Death 1 Receptor , Neoplasms/drug therapy , Signal Transduction , T-Lymphocytes/metabolism , B7-H1 Antigen , Immunotherapy
3.
Acta Biomater ; 161: 184-200, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36893957

ABSTRACT

Non-small cell lung cancer (NSCLC) remains the most frequently diagnosed lung cancer and the leading cause of cancer-related mortality worldwide. PD-1/PD-L1 axis inhibitors have changed the treatment paradigm for various cancer types, including NSCLC. However, success of these inhibitors in lung cancer clinic is severely limited by their inability to inhibit the PD-1/PD-L1 signaling axis due to heavy glycosylation and heterogeneity expression of PD-L1 in NSCLC tumor tissue. Taking advantage of the facts that tumor cell derived nanovesicles could efficiently accumulate in the homotypic tumor sites due to their innate targeting abilities and that specific and high affinity existed between PD-1 and PD-L1, we developed NSCLC targeting biomimetic nanovesicles (NV) cargos from genetically engineered NSCLC cell lines that overexpressed PD-1 (P-NV). We showed that P-NVs efficiently bound NSCLC cells in vitro and targeted tumor nodules in vivo. We further loaded P-NVs with 2-deoxy-D-glucose (2-DG) and doxorubicin (DOX), and found that these drugs co-loaded P-NVs efficiently shrank lung cancers in mouse models for both allograft and autochthonous tumor. Mechanistically, drug-loaded P-NVs efficiently caused cytotoxicity to tumor cells and simultaneously activated anti-tumor immunity function of tumor-infiltrating T cells. Our data therefore strongly argue that 2-DG and DOX co-loaded, PD-1-displaying nanovesicles is a highly promising therapy for treatment of NSCLC in clinic. STATEMENT OF SIGNIFICANCE: Lung cancer cells overexpressing PD-1 are developed for preparing nanoparticles (P-NV). PD-1s displayed on NVs enhance their homologous targeting abilities to tumor cells expressing PD-L1s. Chemotherapeutics such as DOX and 2-DG, are packaged in such nanovesicles (PDG-NV). These nanovesicles efficiently delivered chemotherapeutics to tumor nodules specifically. The synergy between DOX and 2-DG is observed in inhibiting lung cancer cells in vitro and in vivo. Importantly, 2-DG causes deglycosylation and downregulation of PD-L1 on tumor cells while PD-1 displayed on nanovesicles' membrane blocks PD-L1 on tumor cells. 2-DG loaded nanoparticles thus activate anti-tumor activities of T cells in the tumor microenvironment. Our work thus highlights the promising antitumor activity of PDG-NVs, which warrants further clinical evaluation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/pathology , B7-H1 Antigen , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/therapeutic use , Immunotherapy , Doxorubicin/therapeutic use , Cell Line, Tumor , Tumor Microenvironment
4.
Cell Biosci ; 12(1): 193, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36457047

ABSTRACT

Systemic identification of tumor suppressor genes (TSGs) and elucidation of their signaling provide a new angle for understanding of tumorigenesis, which is important for developing successful treatment for lung cancer patients. In our current work, we conducted an in vivo screen for lung cancer TSGs through CRISPR/Cas9 mediated knockout of genes at genome-wide scale. We found that ZNF24 was a potent and clinically relevant TSG of lung cancer. Ectopic expression of ZNF24 arrested lung cancer cells in S phase. Mechanistically, ZNF24 bound to promoter region of P65 to negatively regulate its transcription and thereby the signaling activity of NF-κB pathway. This signaling cascade is clinically relevant. Importantly, we found that combinational inhibition of KRAS, NF-κB, and PD-1 effectively shrank autochthonous KrasG12D/ZNF24-/- lung cancers in transgenic mouse model. Our current work thus revealed an important role played by loss of function of ZNF24 in lung tumorigenesis and shed new light in precision medicine for a portion of lung cancer patients.

5.
Theranostics ; 11(13): 6592-6606, 2021.
Article in English | MEDLINE | ID: mdl-33995678

ABSTRACT

Purpose: Clinical success of cancer therapy is severely limited by drug resistance, attributed in large part to the loss of function of tumor suppressor genes (TSGs). Developing effective strategies to treat those tumors is challenging, but urgently needed in clinic. Experimental Design: MYOCD is a clinically relevant TSG in lung cancer patients. Our in vitro and in vivo data confirm its tumor suppressive function. Further analysis reveals that MYOCD potently inhibits stemness of lung cancer stem cells. Mechanistically, MYOCD localizes to TGFBR2 promoter region and thereby recruits PRMT5/MEP50 complex to epigenetically silence its transcription. Conclusions: NSCLC cells deficient of MYOCD are particularly sensitive to TGFBR kinase inhibitor (TGFBRi). TGFBRi and stemness inhibitor synergize with existing drugs to treat MYOCD deficient lung cancers. Our current work shows that loss of function of MYOCD creates Achilles' heels in lung cancer cells, which might be exploited in clinic.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Nuclear Proteins/deficiency , Receptor, Transforming Growth Factor-beta Type II/antagonists & inhibitors , Trans-Activators/deficiency , Adaptor Proteins, Signal Transducing/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Down-Regulation , Drug Synergism , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Code , Humans , Lung Neoplasms/genetics , Methylation , Mice, Transgenic , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplastic Stem Cells/pathology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/physiology , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/physiology , Tumor Burden
6.
Front Microbiol ; 12: 793923, 2021.
Article in English | MEDLINE | ID: mdl-35126332

ABSTRACT

Bacterial antibiotic resistance sets a great challenge to human health. It seems that the bacteria can spontaneously evolve resistance against any antibiotic within a short time without the horizontal transfer of heterologous genes and before accumulating drug-resistant mutations. We have shown that the tRNA-mediated translational regulation counteracts the reactive oxygen species (ROS) in bacteria. In this study, we demonstrated that isolated and subcultured Escherichia coli elevated its tRNAs under antibiotic stress to rapidly provide antibiotic resistance, especially at the early stage, before upregulating the efflux pump and evolving resistance mutations. The DNA recombination system repaired the antibiotic-induced DNA breakage in the genome, causing numerous structural variations. These structural variations are overrepresented near the tRNA genes, which indicated the cause of tRNA up-regulation. Knocking out the recombination system abolished the up-regulation of tRNAs, and coincidently, they could hardly evolve antibiotic resistance in multiple antibiotics, respectively. With these results, we proposed a multi-stage model of bacterial antibiotic resistance in an isolated scenario: the early stage (recombination-tRNA up-regulation-translational regulation); the medium stage (up-regulation of efflux pump); the late stage (resistant mutations). These results also indicated that the bacterial DNA recombination system and tRNA could be targeted to retard the bacterial spontaneous drug resistance.

7.
Theranostics ; 10(25): 11520-11534, 2020.
Article in English | MEDLINE | ID: mdl-33052230

ABSTRACT

Purpose: Clinical success of precision medicine is severely limited by de novo or acquired drug resistance. It remains a clinically unmet need to treat these patients. Tumor suppressor genes (TSGs) play a critical role in tumorigenesis and impact the therapeutic effect of various treatments. Experimental Design: Using clinical data, in vitro cell line data and in vivo mouse model data, we revealed the tumor suppressive role of Clusterin in lung cancer. We also delineated the signaling cascade elicited by loss of function of CLU in NSCLC cells and tested precision medicine for CLU deficient lung cancers. Results:CLU is a potent and clinically relevant TSG in lung cancer. Mechanistically, CLU inhibits TGFBR1 to recruit TRAF6/TAB2/TAK1 complex and thus inhibits activation of TAK1- NF-κB signaling axis. Lung cancer cells with loss of function of CLU show exquisite sensitivity to TAK1 inhibitors. Importantly, we show that a significant portion of Kras mutation positive NSCLC patients are concurrently deficient of CLU and that TAK1 kinase inhibitor synergizes with existing drugs to treat this portion of lung cancers patients. Conclusions: Combinational treatment with TAK1 inhibitor and MEK1/2 inhibitor effectively shrank Kras mutation positive and CLU deficient NSCLC tumors. Moreover, we put forward a concept that loss of function of a TSG rewires signaling network and thereby creates an Achilles' heel in tumor cells which could be exploited in precision medicine.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Clusterin/deficiency , Lung Neoplasms/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Tumor Suppressor Proteins/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Clusterin/genetics , Datasets as Topic , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , Loss of Function Mutation , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Precision Medicine/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
8.
Front Oncol ; 10: 824, 2020.
Article in English | MEDLINE | ID: mdl-32596145

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide. Tumor suppressor genes (TSGs) play a critical role in restricting tumorigenesis and impact the therapeutic effect of various treatments. However, TSGs remain to be systemically determined in lung cancer. Here, we identified GATA6 as a potent lung cancer TSG. GATA6 inhibited lung cancer cell growth in vitro and tumorigenesis in vivo. Mechanistically, GATA6 upregulated p53 and p21 mRNA while it inhibited AKT activation to stabilize p21 protein, thus inducing lung cancer cell senescence. Furthermore, we showed that ectopic expression of GATA6 led to dramatic slowdown of growth rate of established lung tumor xenograft in vivo.

9.
Molecules ; 25(7)2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32268564

ABSTRACT

Aberrant activation of the WNT/ß-catenin signaling pathway is implicated in various types of cancers. Inhibitors targeting the Wnt signaling pathway are intensively studied in the current cancer research field, the outcomes of which remain to be determined. In this study, we have attempted to discover novel potent WNT/ß-catenin pathway inhibitors through tankyrase 1/2 structure-based virtual screening. After screening more than 13.4 million compounds through molecular docking, we experimentally verified one compound, LZZ-02, as the most potent inhibitor out of 11 structurally representative top hits. LiCl-induced HEK293 cells containing TOPFlash reporter showed that LZZ-02 inhibited the transcriptional activity of ß-catenin with an IC50 of 10 ± 1.2 µM. Mechanistically, LZZ-02 degrades the expression of ß-catenin by stabilizing axin 2, thereby diminishing downstream proteins levels, including c-Myc and cyclin D1. LZZ-02 also inhibits the growth of colonic carcinoma cell harboring constitutively active ß-catenin. More importantly, LZZ-02 effectively shrinks tumor xenograft derived from colonic cell lines. Our study successfully identified a novel tankyrase 1/2 inhibitor and shed light on a novel strategy for developing inhibitors targeting the WNT/ß-catenin signaling axis.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Tankyrases/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Protein Structure, Secondary , Proto-Oncogene Proteins c-myb/metabolism , Xenograft Model Antitumor Assays
10.
Theranostics ; 10(2): 484-497, 2020.
Article in English | MEDLINE | ID: mdl-31903133

ABSTRACT

Rationale: Effective targeting therapies are limited in Hepatocellular carcinoma (HCC) clinic. Characterization of tumor suppressor genes (TSGs) and elucidation their signaling cascades could shed light on new strategies for developing targeting therapies for HCC. Methods: We checked genome-wide DNA copy number variation (CNV) of HCC samples, focusing on deleted genes for TSG candidates. Clinical data, in vitro and in vivo data were collected to validate the tumor suppressor functions. Results: Focal deletion of GATA4 gene locus was the most prominent feature across all liver cancer samples. Ectopic expression of GATA4 resulted in senescence of HCC cell lines. Mechanistically, GATA4 exerted tumor suppressive role by orchestrating the assembly of a tumor suppressor enhancing module: GATA4 directly bound and potently inhibited the mRNA transcription activity of ß-catenin; meanwhile, ß-catenin was recruited by GATA4 to promoter regions and facilitated transcription of GATA4 target genes, which were TSGs per se. Expression of GATA4 was effective to shrink GATA4-deficient HCC tumors in vivo. We also showed that ß-catenin inhibitor was capable of shrinking GATA4-deficient tumors. Conclusions: Our study unveiled a previously unnoticed tumor suppressor enhancing module assembled by ectopically expressed GATA4 in HCC cells and denoted a therapeutic opportunity for GATA4 deficient HCC patients. Our study also presented an interesting case that an oncogenic transcription factor conditionally functioned as a tumor suppressor when recruited by a TSG transcription factor.


Subject(s)
Carcinoma, Hepatocellular/pathology , DNA Copy Number Variations , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Liver Neoplasms/pathology , beta Catenin/metabolism , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Female , GATA4 Transcription Factor/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Signal Transduction , Xenograft Model Antitumor Assays
11.
Cell Cycle ; 18(23): 3300-3312, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31594449

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide. However, tumor suppressor genes remain to be systemically determined for lung cancer. Here we report interferon regulatory factor 8 (IRF8), a member of the IRF family of transcription factors, as a potent lung tumor suppressor gene. Expression of IRF8 is frequently diminished in lung tumoral tissues and is associated with prognosis of non-small cell lung cancer (NSCLC) patients. Ectopic expression of IRF8 suppresses the NSCLC cells proliferation in vitro and tumorigenic potential in vivo. More importantly, forced expression of IRF8 through infection of recombinant virus inhibits lung tumorigenesis in genetically engineered mouse model (GEMM). Mechanistically, IRF8 inhibits AKT signaling and promotes accumulation of P27 protein, which results in senescence of lung cancer cells. Ectopic expression of IRF8 in tumor cells leads to regression of lung cancer tumor nodules in a xenograft tumor model. Our data, therefore, solidly shows IRF8 to be a lung cancer suppressor gene and may denote an opportunity for therapeutic intervention of NSCLC.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cellular Senescence/genetics , Interferon Regulatory Factors/genetics , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Mice , Prognosis , Signal Transduction/genetics , Tumor Suppressor Proteins/genetics
12.
Nat Commun ; 10(1): 1665, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30971692

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide. Tumor suppressor genes remain to be systemically identified for lung cancer. Through the genome-wide screening of tumor-suppressive transcription factors, we demonstrate here that GATA4 functions as an essential tumor suppressor in lung cancer in vitro and in vivo. Ectopic GATA4 expression results in lung cancer cell senescence. Mechanistically, GATA4 upregulates multiple miRNAs targeting TGFB2 mRNA and causes ensuing WNT7B downregulation and eventually triggers cell senescence. Decreased GATA4 level in clinical specimens negatively correlates with WNT7B or TGF-ß2 level and is significantly associated with poor prognosis. TGFBR1 inhibitors show synergy with existing therapeutics in treating GATA4-deficient lung cancers in genetically engineered mouse model as well as patient-derived xenograft (PDX) mouse models. Collectively, our work demonstrates that GATA4 functions as a tumor suppressor in lung cancer and targeting the TGF-ß signaling provides a potential way for the treatment of GATA4-deficient lung cancer.


Subject(s)
GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Lung Neoplasms/genetics , Wnt Proteins/metabolism , A549 Cells , Animals , Cellular Senescence/genetics , Down-Regulation , Female , GATA4 Transcription Factor/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Prognosis , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta2/metabolism , Up-Regulation , Wnt Proteins/genetics , Xenograft Model Antitumor Assays
13.
ACS Biomater Sci Eng ; 4(1): 142-150, 2018 Jan 08.
Article in English | MEDLINE | ID: mdl-33418684

ABSTRACT

Fluorescent carbon dots (CDs) as a luminescent nanomaterial have obtained much attention in the biomedical field. To make good use of their luminescent property and nanoscaled size, we developed CDs as a vaccine delivery system for intranasal immunization in this work. To this aim, polyethylenimine-modified CDs were prepared via a simple microwave method. Intranasal immunization was performed by using the CDs as an antigen carrier to deliver model protein antigen ovalbumin. The results showed that the CDs as an intranasal vaccine delivery system enhanced the immunization efficacy by significantly increasing IgG titer, IgA induction in the local and distant mucous membrane sites, splenocyte proliferation, cytokine IFN-γ secretion by splenocytes, and memory T cells. From the results, the CDs could be used as vaccine delivery systems with the advantage of tracing the antigen transportation from administration site to the lymph organs.

14.
J Inorg Biochem ; 167: 150-156, 2017 02.
Article in English | MEDLINE | ID: mdl-28341101

ABSTRACT

The pathogenic Streptococcus pneumoniae (S. pneumoniae) has evolved a special mechanism such as pneumococcal iron acquisition ATP binding cassette (PiaABC) to take up siderophore-iron from its host. The cell-surface lipoprotein PiaA, a key component of PiaABC, is the primary receptor to bind ferrichrome (Fc). To study the structure-function relationship of PiaA, three conservative amino-acid residues, Trp63, Trp158 and Phe255, in the hydrophobic barrel of the metal binding site of PiaA, were individually and collectively mutated to alanine; and the resulted single-point mutants, W63A, W158A and F255A, and triple mutant W63A/W158A/F255A were characterized by using biochemical and biophysical methods. Experiments showed that wild-type PiaA (WT-PiaA) and the single-point mutant proteins bound Fc with a similar kinetics mode, but the reaction rate of W158A was lower than that for WT-PiaA. The binding affinity of W158A toward Fc was significantly weaker than that of the WT-PiaA-Fc (wild-type PiaA bound with Fc) interaction. Furthermore, the absence of Trp158 in the protein led to a significant impact on the secondary structure of PiaA, resulting in a labile conformational structure of W158A, with impaired resistance to thermal and chemical denaturation. Collectively, Trp158 is a crucial residue for binding Fc, playing an important role in stabilizing the PiaA-Fc complex. This study revealed the critical role of the conserved tryptophan residues in Fc-binding protein PiaA, and provided valuable information for understanding the Fc transport mechanism mediated by PiaA or its homologous proteins in bacteria.


Subject(s)
Bacterial Proteins/chemistry , Ferrichrome/chemistry , Lipoproteins/chemistry , Multiprotein Complexes/chemistry , Streptococcus pneumoniae/chemistry , Amino Acid Substitution , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ferrichrome/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation, Missense , Protein Binding , Protein Denaturation , Protein Structure, Quaternary , Protein Structure, Secondary , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Tryptophan/chemistry , Tryptophan/genetics , Tryptophan/metabolism
16.
Metallomics ; 7(3): 448-54, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25608595

ABSTRACT

Streptococcus pneumoniae is a Gram-positive bacterial pathogen causing a variety of diseases, including otitis media, bacteraemia and meningitis. Although copper is an essential trace metal for bacterial growth, high intracellular levels of free-copper are toxic. Copper resistance has emerged as an important virulence determinant of microbial pathogens. In this study, we determined the minimum inhibition concentration of copper for the growth inhibition of S. pneumoniae. Two-dimensional-electrophoresis coupled with mass spectrometry was applied to identify proteins involved in copper resistance of S. pneumoniae. In total, forty-four proteins with more than 1.5-fold alteration in expression (p < 0.05) were identified. Quantitative reverse transcription PCR was used to confirm the proteomic results. Bioinformatics analysis showed that the differentially expressed proteins were mainly involved in the cell wall biosynthesis, protein biosynthesis, purine biosynthesis, pyrimidine biosynthesis, primary metabolic process, and the nitrogen compound metabolic process. Many up-regulated proteins in response to the copper treatment directly or indirectly participated in the cell wall biosynthesis, indicating that the cell wall is a critical determinant in copper resistance of S. pneumoniae.


Subject(s)
Copper/pharmacology , Proteomics/methods , Streptococcus pneumoniae/metabolism , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Wall/drug effects , Cell Wall/metabolism , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Bacterial/drug effects , Genes, Bacterial , Intracellular Space/metabolism , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development
17.
J Proteomics ; 115: 107-16, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25497219

ABSTRACT

Streptococcus pneumoniae is a Gram-positive pathogen that causes a variety of infection diseases in human. In this project, we determined the antibacterial activity of a Ru(II) complex X-03 against S. pneumoniae in vitro, by comparing its toxicity to host cells A549 and HBE. We performed two-dimensional gel electrophoresis (2-DE)-based proteomic analysis to characterize the protein alterations in S. pneumoniae after treatment with X-03. In total, 50 proteins exhibiting significant differential expressions were identified. RT-PCR was used to confirm the expression differences for selected proteins. Bioinformatics analysis on the proteomic alterations suggested that Ru(II) complex X-03 may obstruct bacterial fatty acid synthesis and oxidation-reduction process to suppress the growth of S. pneumoniae. Metal-uptake experiments revealed that iron-acquisition pathway in the bacterium may be interfered by X-03. These results provide useful clues for further investigations on the mechanism of the antibacterial action of metal compounds. BIOLOGICAL SIGNIFICANCE: The appearance of bacterial strains with broad antibiotic resistance is becoming an alarming global health concern. The development of novel efficient antibacterial compound is urgently needed. In the present study, we found that Ru(II) complex X-03 has a significant antibacterial activity and applied proteomic technology combined with bioinformatics analysis to investigate its antimicrobial mechanism in S. pneumoniae. Many proteins were found to be dysregulated, implicating that X-03 may affect various molecular pathways leading to the inhibition of bacterial growth. Metal-uptake experiments demonstrated that X-03 treatment reduced the iron content in the bacterium, suggesting the interference with iron acquisition systems by the complex. This disturbance in iron acquisition may directly or indirectly induce the proteomic response that involved many pathways. In addition, X-03 could selectively suppress Gram-positive bacteria but execute less cytotoxicity to Gram-negative bacteria, with almost no effect on human cells, implicating its potential to be developed as a specific antimicrobial agent. These results provide useful information for further investigations on the mechanism of the antibacterial action of metal drugs and development of efficient antibacterial drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Ruthenium/pharmacology , Streptococcus pneumoniae/metabolism , Anti-Bacterial Agents/chemistry , Cell Line , Humans , Proteomics , Ruthenium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...