Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Genet ; 19(12): e1011098, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38134213

ABSTRACT

Cell death resistance is a hallmark of tumor cells that drives tumorigenesis and drug resistance. Targeting cell death resistance-related genes to sensitize tumor cells and decrease their cell death threshold has attracted attention as a potential antitumor therapeutic strategy. However, the underlying mechanism is not fully understood. Recent studies have reported that NeuroD1, first discovered as a neurodifferentiation factor, is upregulated in various tumor cells and plays a crucial role in tumorigenesis. However, its involvement in tumor cell death resistance remains unknown. Here, we found that NeuroD1 was highly expressed in hepatocellular carcinoma (HCC) cells and was associated with tumor cell death resistance. We revealed that NeuroD1 enhanced HCC cell resistance to ferroptosis, a type of cell death caused by aberrant redox homeostasis that induces lipid peroxide accumulation, leading to increased HCC cell viability. NeuroD1 binds to the promoter of glutathione peroxidase 4 (GPX4), a key reductant that suppresses ferroptosis by reducing lipid peroxide, and activates its transcriptional activity, resulting in decreased lipid peroxide and ferroptosis. Subsequently, we showed that NeuroD1/GPX4-mediated ferroptosis resistance was crucial for HCC cell tumorigenic potential. These findings not only identify NeuroD1 as a regulator of tumor cell ferroptosis resistance but also reveal a novel molecular mechanism underlying the oncogenic function of NeuroD1. Furthermore, our findings suggest the potential of targeting NeuroD1 in antitumor therapy.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Lipid Peroxides , Ferroptosis/genetics , Liver Neoplasms/genetics , Peroxides , Carcinogenesis , Cell Line, Tumor
2.
J Genet Genomics ; 48(11): 1020-1031, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34696992

ABSTRACT

L-Arginine is the precursor of nitric oxide (NO), a host immune effector against intracellular pathogens including Mycobacterium tuberculosis (M. tb). Pathogens including M. tb have evolved various strategies targeting arginine to block the production of NO for better survival and proliferation. However, L-arginine metabolism and regulation in Mycobacterium are poorly understood. Here, we report the identification of M. smegmatis MSMEG_1415 (homolog of M. tb Rv2324) as an arginine-responsive transcriptional factor regulating the arginase pathway. In the absence of L-arginine, MSMEG_1415 acts as a repressor to inhibit the transcription of the roc (for arginine, ornithine catabolism) gene cluster, thereby switching off the arginase pathway. Treatment with L-arginine relieves the transcriptional inhibition of MSMEG_1415 on the roc gene cluster to activate the arginase pathway. Moreover, the L-arginine-MSMEG_1415 complex activates the transcription of the roc gene cluster by recognizing and binding a 15-bp palindrome motif, thereby preventing the excess accumulation of L-arginine in M. smegmatis. Physiologically, MSMEG_1415 confers mycobacteria resistance to starvation and fluoroquinolones exposure, suggestive of its important role in M. smegmatis persistence. The results uncover a unique regulatory mechanism of arginine metabolism in mycobacteria and identify M. tb Rv2324 as an attractive candidate target for the design of drugs against tuberculosis.


Subject(s)
Arginase/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Mycobacterium Infections/microbiology , Mycobacterium/metabolism , Transcription Factors/metabolism , Bacterial Proteins/genetics , Metabolic Networks and Pathways , Multigene Family , Promoter Regions, Genetic , Sequence Deletion , Transcription Factors/genetics
3.
Appl Microbiol Biotechnol ; 104(5): 2137-2147, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31940082

ABSTRACT

Tuberculosis caused by Mycobacterium tuberculosis remains a major global health concern; M. tuberculosis drug resistance and persistence further fueled the situation. Nutrient supportive therapy was intensively pursued to complement the conventional treatment, as well as their synergy with current antibiotics. To explore whether L-alanine can synergize with fluoroquinolones against M. tuberculosis, M. smegmatis was used as a surrogate in this study. We found that L-alanine can boost the bactericidal efficacy of fluoroquinolones, increasing the production of intracellular reactive oxygen species. This effect is very significant for persisters. Accelerated tricarboxylic acid cycle and/or nucleotide metabolism were observed after the addition of L-alanine. M. smegmatis MSMEG2660 is a homolog of the alanine dehydrogenase (Rv2780, MSMEG2659) negative regulator Rv2779c and involved in the L-alanine potentiation of fluoroquinolone via funneling more alanine into tricarboxylic acid. Deletion mutant of the MSMEG2660 (∆Ms2660) became more susceptible, and more readily revived from persistence. We firstly found that L-alanine can synergize with fluoroquinolones against Mycobacterium, especially the persisters via promoting metabolism. This will inspire new avenue to eliminate Mycobacterium persisters.


Subject(s)
Alanine/metabolism , Anti-Bacterial Agents/pharmacology , Fluoroquinolones/pharmacology , Mycobacterium smegmatis/drug effects , Mycobacterium tuberculosis/drug effects , Reactive Oxygen Species/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Oxidative Stress/drug effects
4.
Infect Genet Evol ; 77: 104070, 2020 01.
Article in English | MEDLINE | ID: mdl-31614213

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) is still a leading cause of death worldwide. M. tuberculosis has evolved multipronged strategies to subvert host immune defenses and establish an immunologically privileged niche in macrophages. Rv0426c has been predicted to be an effector involved in the Mtb-host interactions. To investigate the potential role played by Rv0426c, we constructed recombinant M. smegmatis strains with heterologous expression of Rv0426c. We observed that Rv0426c recombinants became more susceptible to various stresses by increasing cell wall permeability, however with elevated early survival rate within macrophages. This was accompanied by decreased levels of pro-inflammatory cytokines and host cell apoptosis. The data suggested that Rv0426c was a new player involved in the interactions between Mtb and macrophages.


Subject(s)
Bacterial Proteins/metabolism , Macrophages/cytology , Mycobacterium smegmatis/physiology , Mycobacterium tuberculosis/metabolism , Apoptosis , Bacterial Proteins/genetics , Cell Differentiation/drug effects , Cell Wall/metabolism , Cytokines/metabolism , Down-Regulation , Host-Pathogen Interactions , Humans , Macrophages/immunology , Macrophages/microbiology , Microbial Viability , Mycobacterium smegmatis/genetics , Recombinant Proteins/metabolism , THP-1 Cells , Tetradecanoylphorbol Acetate/pharmacology
5.
Arch Biochem Biophys ; 667: 59-66, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31054279

ABSTRACT

Development of extensively drug resistant (XDR) strains and multidrug resistant (MDR) in Mycobacterium tuberculosis is caused by an efflux mechanism of antibiotics in the bacteria. Rv0191, predicted to a major facilitator superfamily transporter of efflux pump, contributes to elevated expression in some clinical isolates. To characterize the role of Rv0191 which might be involved in antibiotics resistance, Mycobacterium smegmatis was taken as a type strains to do drug susceptibility, ethidium bromide (EB) accumulation assay and electrophoretic mobility shift assay. M. smegmatis Ms0232 mutant became more susceptible to chloramphenicol and showed different cell surface properties. Rv1353c, a TetR family transcription factor, can downregulate the transcription of Rv0191. Rv1353c overexpression strain became more sensitive to chloramphenicol. Together, these findings indicate that Rv1353c encodes a transcriptional repressor that directly interacts with the Rv0191 promoter and modulates the expression of Rv0191. This provided a new player in mycobacteria chloramphenicol resistance.


Subject(s)
Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Bacterial Proteins/genetics , Chloramphenicol/pharmacokinetics , Chloramphenicol/pharmacology , Drug Resistance, Bacterial/genetics , Gene Expression Regulation, Bacterial , Gene Knockout Techniques , Genes, Bacterial , Humans , Membrane Transport Proteins/genetics , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Promoter Regions, Genetic
6.
J Cell Biochem ; 120(8): 12197-12207, 2019 08.
Article in English | MEDLINE | ID: mdl-31026098

ABSTRACT

The typical two-component regulatory systems (TCSs), consisting of response regulator and histidine kinase, play a central role in survival of pathogenic bacteria under stress conditions such as nutrient starvation, hypoxia, and nitrosative stress. A total of 11 complete paired two-component regulatory systems have been found in Mycobacterium tuberculosis, including a few isolated kinase and regulatory genes. Increasing evidence has shown that TCSs are closely associated with multiple physiological process like intracellular persistence, pathogenicity, and metabolism. This review gives the two-component signal transduction systems in M. tuberculosis and their signal transduction roles in adaption to the environment.


Subject(s)
Microbial Viability , Mycobacterium tuberculosis/metabolism , Signal Transduction , Models, Biological , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/microbiology , Virulence Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...