1
|
Paroha R, Wang J, Lee S. PDCD4 as a marker of mTOR pathway activation and therapeutic target in mycobacterial infections. Microbiol Spectr 2024; 12:e0006224. [PMID: 38912807 PMCID: PMC11302300 DOI: 10.1128/spectrum.00062-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Programmed cell death protein 4 (PDCD4) is instrumental in regulating a range of cellular processes such as translation, apoptosis, signal transduction, and inflammatory responses. There is a notable inverse correlation between PDCD4 and the mammalian target of rapamycin (mTOR) pathway, which is integral to cellular growth control. Activation of mTOR is associated with the degradation of PDCD4. Although the role of PDCD4 is well established in oncogenesis and immune response regulation, its function in mycobacterial infections and its interplay with the mTOR pathway necessitate further elucidation. This study investigates the modulation of PDCD4 expression in the context of mycobacterial infections, revealing a consistent pattern of downregulation across diverse mycobacterial species. This observation underscores the potential utility of PDCD4 as a biomarker for assessing mTOR pathway activation during such infections. Building on this finding, we employed a novel approach using PDCD4-based mTOR (Tor)-signal-indicator (TOSI) reporter cells for the high-throughput screening of FDA-approved drugs, focusing on mTOR inhibitors. This methodology facilitated the identification of several agents, inclusive of known mTOR inhibitors, which upregulated PDCD4 expression and concurrently exhibited efficacy in impeding mycobacterial proliferation within macrophages. These results not only reinforce the significance of PDCD4 as a pivotal marker in the understanding of infectious diseases, particularly mycobacterial infections, but also illuminate its potential in the identification of mTOR inhibitors, thereby contributing to the advancement of therapeutic strategies. IMPORTANCE This study emphasizes the critical role of the mammalian target of rapamycin (mTOR) pathway in macrophage responses to mycobacterial infections, elucidating how mycobacteria activate mTOR, resulting in PDCD4 degradation. The utilization of the (Tor)-signal-indicator (TOSI) vector for real-time monitoring of mTOR activity represents a significant advancement in understanding mTOR regulation during mycobacterial infection. These findings deepen our comprehension of mycobacteria's innate immune mechanisms and introduce PDCD4 as a novel marker for mTOR activity in infectious diseases. Importantly, this research laid the groundwork for high-throughput screening of mTOR inhibitors using FDA-approved drugs, offering the potential for repurposing treatments against mycobacterial infections. The identification of drugs that inhibit mTOR activation opens new avenues for host-directed therapies, marking a significant step forward in combating tuberculosis and other mycobacterial diseases.
Collapse
Affiliation(s)
- Ruchi Paroha
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Jia Wang
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Sunhee Lee
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
2
|
Fayed B, Shakartalla SB, Sabbah H, Dalle H, Tannira M, Senok A, Soliman SSM. Transcriptome Analysis of Human Dermal Cells Infected with Candida auris Identified Unique Pathogenesis/Defensive Mechanisms Particularly Ferroptosis. Mycopathologia 2024; 189:65. [PMID: 38990436 DOI: 10.1007/s11046-024-00868-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
Candida auris is an emerging multi-drug resistant yeast that can cause life-threatening infections. A recent report clarified the ability of C. auris to form a biofilm with enhanced drug resistance properties in the host skin's deep layers. The formed biofilm may initiate further bloodstream spread and immune escape. Therefore, we propose that secreted chemicals from the biofilm may facilitate fungal pathogenesis. In response to this interaction, the host skin may develop potential defensive mechanisms. Comparative transcriptomics was performed on the host dermal cells in response to indirect interaction with C. auris biofilm through Transwell inserts compared to planktonic cells. Furthermore, the effect of antifungals including caspofungin and fluconazole was studied. The obtained data showed that the dermal cells exhibited different transcriptional responses. Kyoto Encyclopedia of Genes and Genomes and Reactome analyses identified potential defensive responses employed by the dermal cells and potential toxicity induced by C. auris. Additionally, our data indicated that the dominating toxic effect was mediated by ferroptosis; which was validated by qRT-PCR, cytotoxicity assay, and flow cytometry. On the other hand, the viability of C. auris biofilm was enhanced and accompanied by upregulation of MDR1, and KRE6 upon interaction with dermal cells; both genes play significant roles in drug resistance and biofilm maturation, respectively. This study for the first-time shed light on the dominating defensive responses of human dermal cells, microbe colonization site, to C. auris biofilm and its toxic effects. Further, it demonstrates how C. auris biofilm responds to the defensive mechanisms developed by the human dermal cells.
Collapse
Affiliation(s)
- Bahgat Fayed
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Chemistry of Natural and Microbial Products, National Research Centre, Cairo, Egypt
| | - Sarra B Shakartalla
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Faculty of Pharmacy, University of Gezira, P.O.Box. 21111, Wad Medani, Sudan
| | - Hassan Sabbah
- AbbVie BioPharmaceuticals, P.O. Box 118052, Dubai, UAE
| | - Hala Dalle
- AbbVie BioPharmaceuticals, Kuwait City, Kuwait
| | | | - Abiola Senok
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14 Dubai Healthcare City, P.O.Box 505055, Dubai, UAE
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
| |
Collapse
|
3
|
Zhang W, Dong C, Xiong S. Mycobacterial SapM hampers host autophagy initiation for intracellular bacillary survival via dephosphorylating Raptor. iScience 2024; 27:109671. [PMID: 38646170 PMCID: PMC11031826 DOI: 10.1016/j.isci.2024.109671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/01/2024] [Accepted: 04/02/2024] [Indexed: 04/23/2024] Open
Abstract
Secreted acid phosphatase (SapM) is an immunomodulator of Mycobacterium tuberculosis (Mtb) and consequently plays a crucial role in disease onset and development upon infection. Importantly, the virulence of SapM has rendered SapM an attractive target for drug development. However, the mechanism underlying the role of SapM in facilitating bacillary survival remains to be fully elucidated. In this context, the present study demonstrated that SapM hampered cellular autophagy to facilitate bacillary survival in mycobacterial-infected macrophages. Mechanically, SapM interacted with Raptor and was localized to the subcellular lysosomal organelle, causing the dephosphorylation of Raptor at the Ser792 position, resulting in mTORC1 hyperactivity and the subsequent autophagy inhibition. Consistent with this, SapM blocked the autophagy initiation and mitigated lung pathology in vivo. These findings highlighted the role of Raptor as a significant substrate of SapM for inhibiting autophagy, which is a novel clue for developing a treatment against tuberculosis.
Collapse
Affiliation(s)
- Wei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Chunsheng Dong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
- Key Laboratory of Geriatric Diseases and Immunology, Ministry of Education, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
4
|
Bhatnagar A, Chopra U, Raja S, Das KD, Mahalingam S, Chakravortty D, Srinivasula SM. TLR-mediated aggresome-like induced structures comprise antimicrobial peptides and attenuate intracellular bacterial survival. Mol Biol Cell 2024; 35:ar34. [PMID: 38170582 PMCID: PMC10916861 DOI: 10.1091/mbc.e23-09-0347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Immune cells employ diverse mechanisms for host defense. Macrophages, in response to TLR activation, assemble aggresome-like induced structures (ALIS). Our group has shown TLR4-signaling transcriptionally upregulates p62/sequestome1, which assembles ALIS. We have demonstrated that TLR4-mediated autophagy is, in fact, selective-autophagy of ALIS. We hypothesize that TLR-mediated autophagy and ALIS contribute to host-defense. Here we show that ALIS are assembled in macrophages upon exposure to different bacteria. These structures are associated with pathogen-containing phagosomes. Importantly, we present evidence of increased bacterial burden, where ALIS assembly is prevented with p62-specific siRNA. We have employed 3D-super-resolution structured illumination microscopy (3D-SR-SIM) and mass-spectrometric (MS) analyses to gain insight into the assembly of ALIS. Ultra-structural analyses of known constituents of ALIS (p62, ubiquitin, LC3) reveal that ALIS are organized structures with distinct patterns of alignment. Furthermore, MS-analyses of ALIS identified, among others, several proteins of known antimicrobial properties. We have validated MS data by testing the association of some of these molecules (Bst2, IFITM2, IFITM3) with ALIS and the phagocytosed-bacteria. We surmise that AMPs enrichment in ALIS leads to their delivery to bacteria-containing phagosomes and restricts the bacteria. Our findings in this paper support hitherto unknown functions of ALIS in host-defense.
Collapse
Affiliation(s)
- Anushree Bhatnagar
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Umesh Chopra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sebastian Raja
- Laboratory of Molecular Cell Biology, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India
| | - Krishanu Dey Das
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - S. Mahalingam
- Laboratory of Molecular Cell Biology, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India
| | - Dipshikha Chakravortty
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Srinivasa Murty Srinivasula
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| |
Collapse
|
5
|
Abdalla AE, Alanazi A, Abosalif KOA, Alameen AAM, Junaid K, Manni E, Talha AA, Ejaz H. MicroRNA-155, a double-blade sword regulator of innate tuberculosis immunity. Microb Pathog 2023; 185:106438. [PMID: 37925110 DOI: 10.1016/j.micpath.2023.106438] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/06/2023]
Abstract
Tuberculosis (TB) is a chronic, life-threatening disease caused by unusual facultative intracellular bacteria, Mycobacterium tuberculosis. This bacterium has unique resistance to many antimicrobial agents and has become a major global health concern due to emerging multidrug-resistant strains. Additionally, it has developed multiple schemes to exploit host immune signaling and establish long-term survival within host tissues. Thus, understanding the pathways that govern the crosstalk between the bacterium and the immune system could provide a new avenue for therapeutic interventions. MicroRNAs (miRs) are short, noncoding, and regulator RNA molecules that control the expression of cellular genes by targeting their mRNAs post-transcriptionally. MiR-155 is one of the most crucial miR in shaping the host immune defenses against M. tuberculosis. MiR-155 is remarkably downregulated in patients with clear clinical TB symptoms in comparison with latently infected patients and/or healthy individuals, thereby implicating its role in controlling M. tuberculosis infection. However, functional probing of miR-155 suggests dual effects in regulating the host's innate defenses in response to mycobacterial infection. This review provides comprehensive knowledge and future perspectives regarding complex signaling pathways that mediated miR-155 expression during M. tuberculosis infections. Moreover, miR-155-targeting signaling orchestrates inflammatory mediators' production, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Abualgasim Elgaili Abdalla
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia.
| | - Awadh Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Khalid Omer Abdalla Abosalif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Ayman Ali Mohammed Alameen
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Kashaf Junaid
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Emad Manni
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Albadawi Abdelbagi Talha
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia.
| |
Collapse
|
6
|
Deng F, Xu P, Miao J, Jin C, Tu H, Zhang J. Pulmonary tuberculosis biomarker miR-215-5p inhibits autophagosome-lysosome fusion in macrophages. Tuberculosis (Edinb) 2023; 143:102422. [PMID: 37925930 DOI: 10.1016/j.tube.2023.102422] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/17/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
The normal autophagy flux is beneficial for the rapid elimination of phagocytic pathogens by macrophages. However, Mycobacterium tuberculosis interferes with the autophagy flux of macrophages to weaken their immune function and evade immune surveillance. In this study, we found that miRNA-215-5p was downregulated in tuberculosis patients. A potential diagnostic model for tuberculosis was established by combining miRNA-215-5p with three others differentially expressed microRNAs (miRNA-145-5p, miRNA-486-5p and miRNA-628-3p). Furthermore, we discovered that the up-regulated miRNA-215-5p could inhibit the maturation of autophagy by preventing the fusion of autophagosomes with lysosomes in macrophages. The role of TB-specific miRNA-215-5p in inhibiting auto-lysosome formation provides evidence of its potential role in Host-directed therapy for tuberculosis.
Collapse
Affiliation(s)
- Feng Deng
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Peng Xu
- Department of Clinical Laboratory, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Jiahong Miao
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Cheng Jin
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Huihui Tu
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China.
| | - Jianhua Zhang
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China.
| |
Collapse
|
7
|
Sengupta S, Pattanaik KP, Mishra S, Sonawane A. Epigenetic orchestration of host immune defences by Mycobacterium tuberculosis. Microbiol Res 2023; 273:127400. [PMID: 37196490 DOI: 10.1016/j.micres.2023.127400] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/09/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023]
Abstract
Being among the top 10 causes of adult deaths, tuberculosis (TB) disease is considered a major global public health concern to address. The human tuberculosis pathogen, Mycobacterium tuberculosis (Mtb), is an extremely competent and well-versed pathogen that promotes pathogenesis by evading the host immune systems through numerous tactics. Investigations revealed that Mtb could evade the host defense mechanisms by reconfiguring the host gene transcription and causing epigenetic changes. Although results indicate the link between epigenetics and disease manifestation in other bacterial infections, little is known regarding the kinetics of the epigenetic alterations in mycobacterial infection. This literature review discusses the studies in Mtb-induced epigenetic alterations inside the host and its contribution in the host immune evasion strategies. It also discusses how the Mtb-induced alterations could be used as 'epibiomarkers' to diagnose TB. Additionally, this review also discusses therapeutic interventions to be enhanced through remodification by 'epidrugs'.
Collapse
Affiliation(s)
- Srabasti Sengupta
- School of Biotechnology, Campus-11, KIIT Deemed to be University, Patia, Bhubaneswar 751024, India
| | - Kali Prasad Pattanaik
- School of Biotechnology, Campus-11, KIIT Deemed to be University, Patia, Bhubaneswar 751024, India
| | - Snehasish Mishra
- School of Biotechnology, Campus-11, KIIT Deemed to be University, Patia, Bhubaneswar 751024, India
| | - Avinash Sonawane
- Discipline of Biosciences and Biomedical Engineering, Indian Institutes of Technology Indore, Khandwa Road, Simrol, Indore 453552, India.
| |
Collapse
|
8
|
Alizadeh J, Kavoosi M, Singh N, Lorzadeh S, Ravandi A, Kidane B, Ahmed N, Mraiche F, Mowat MR, Ghavami S. Regulation of Autophagy via Carbohydrate and Lipid Metabolism in Cancer. Cancers (Basel) 2023; 15:2195. [PMID: 37190124 PMCID: PMC10136996 DOI: 10.3390/cancers15082195] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 05/17/2023] Open
Abstract
Metabolic changes are an important component of tumor cell progression. Tumor cells adapt to environmental stresses via changes to carbohydrate and lipid metabolism. Autophagy, a physiological process in mammalian cells that digests damaged organelles and misfolded proteins via lysosomal degradation, is closely associated with metabolism in mammalian cells, acting as a meter of cellular ATP levels. In this review, we discuss the changes in glycolytic and lipid biosynthetic pathways in mammalian cells and their impact on carcinogenesis via the autophagy pathway. In addition, we discuss the impact of these metabolic pathways on autophagy in lung cancer.
Collapse
Affiliation(s)
- Javad Alizadeh
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
| | - Mahboubeh Kavoosi
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
| | - Navjit Singh
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
| | - Amir Ravandi
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Institute of Cardiovascular Sciences, Albrechtsen Research Centre, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
| | - Biniam Kidane
- Section of Thoracic Surgery, Department of Surgery, Health Sciences Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 6C5, Canada;
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (N.A.)
| | - Naseer Ahmed
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (N.A.)
- Department of Radiology, Section of Radiation Oncology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar;
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Michael R. Mowat
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (N.A.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
- Research Institute of Oncology and Hematology, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine in Zabrze, Academia of Silesia, 41-800 Zabrze, Poland
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
| |
Collapse
|
9
|
Kim YJ, Park EJ, Lee SH, Silwal P, Kim JK, Yang JS, Whang J, Jang J, Kim JM, Jo EK. Dimethyl itaconate is effective in host-directed antimicrobial responses against mycobacterial infections through multifaceted innate immune pathways. Cell Biosci 2023; 13:49. [PMID: 36882813 PMCID: PMC9993662 DOI: 10.1186/s13578-023-00992-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Itaconate, a crucial immunometabolite, plays a critical role in linking immune and metabolic functions to influence host defense and inflammation. Due to its polar structure, the esterified cell-permeable derivatives of itaconate are being developed to provide therapeutic opportunities in infectious and inflammatory diseases. Yet, it remains largely uncharacterized whether itaconate derivatives have potentials in promoting host-directed therapeutics (HDT) against mycobacterial infections. Here, we report dimethyl itaconate (DMI) as the promising candidate for HDT against both Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria by orchestrating multiple innate immune programs. RESULTS DMI per se has low bactericidal activity against Mtb, M. bovis Bacillus Calmette-Guérin (BCG), and M. avium (Mav). However, DMI robustly activated intracellular elimination of multiple mycobacterial strains (Mtb, BCG, Mav, and even to multidrug-resistant Mtb) in macrophages and in vivo. DMI significantly suppressed the production of interleukin-6 and -10, whereas it enhanced autophagy and phagosomal maturation, during Mtb infection. DMI-mediated autophagy partly contributed to antimicrobial host defenses in macrophages. Moreover, DMI significantly downregulated the activation of signal transducer and activator of transcription 3 signaling during infection with Mtb, BCG, and Mav. CONCLUSION Together, DMI has potent anti-mycobacterial activities in macrophages and in vivo through promoting multifaceted ways for innate host defenses. DMI may bring light to new candidate for HDT against Mtb and nontuberculous mycobacteria, both of which infections are often intractable with antibiotic resistance.
Collapse
Affiliation(s)
- Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun-Jin Park
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Chungbuk, South Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Jeong Seong Yang
- Department of Research and Development, Korea Mycobacterium Resource Center (KMRC), The Korean Institute of Tuberculosis, Osong, 28158, South Korea
| | - Jake Whang
- Department of Research and Development, Korea Mycobacterium Resource Center (KMRC), The Korean Institute of Tuberculosis, Osong, 28158, South Korea
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, 52828, South Korea
| | - Jin-Man Kim
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Pathology, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea. .,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea. .,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.
| |
Collapse
|
10
|
BORC complex specific components and Kinesin-1 mediate autophagy evasion by the autophagy-resistant Mycobacterium tuberculosis Beijing strain. Sci Rep 2023; 13:1663. [PMID: 36717601 PMCID: PMC9886903 DOI: 10.1038/s41598-023-28983-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Autophagy induction by starvation has been shown to enhance lysosomal delivery to mycobacterial phagosomes, resulting in the restriction of the Mycobacterium tuberculosis reference strain H37Rv. In contrast to H37Rv, our previous study showed that strains belonging to the notorious M. tuberculosis Beijing genotype could evade autophagic elimination. Our recent RNA-Seq analysis also discovered that the autophagy-resistant M. tuberculosis Beijing strain (BJN) evaded autophagic control by upregulating the expression of Kxd1, a BORC complex component, and Plekhm2, both of which function in lysosome positioning towards the cell periphery in host macrophages, thereby suppressing enhanced lysosomal delivery to its phagosome and sparing the BJN from elimination as a result. In this work, we further characterised the other specific components of the BORC complex, BORC5-8, and Kinesin proteins in autophagy resistance by the BJN. Depletion of BORCS5-8 and Kinesin-1, but not Kinesin-3, reverted autophagy avoidance by the BJN, resulting in increased lysosomal delivery to the BJN phagosomes. In addition, the augmented lysosome relocation towards the perinuclear region could now be observed in the BJN-infected host cells depleted in BORCS5-8 and Kinesin-1 expressions. Taken together, the data uncovered new roles for BORCS5-8 and Kinesin-1 in autophagy evasion by the BJN.
Collapse
|
11
|
Bo H, Moure UAE, Yang Y, Pan J, Li L, Wang M, Ke X, Cui H. Mycobacterium tuberculosis-macrophage interaction: Molecular updates. Front Cell Infect Microbiol 2023; 13:1062963. [PMID: 36936766 PMCID: PMC10020944 DOI: 10.3389/fcimb.2023.1062963] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of Tuberculosis (TB), remains a pathogen of great interest on a global scale. This airborne pathogen affects the lungs, where it interacts with macrophages. Acidic pH, oxidative and nitrosative stressors, and food restrictions make the macrophage's internal milieu unfriendly to foreign bodies. Mtb subverts the host immune system and causes infection due to its genetic arsenal and secreted effector proteins. In vivo and in vitro research have examined Mtb-host macrophage interaction. This interaction is a crucial stage in Mtb infection because lung macrophages are the first immune cells Mtb encounters in the host. This review summarizes Mtb effectors that interact with macrophages. It also examines how macrophages control and eliminate Mtb and how Mtb manipulates macrophage defense mechanisms for its own survival. Understanding these mechanisms is crucial for TB prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Haotian Bo
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Ulrich Aymard Ekomi Moure
- The Ninth People's Hospital of Chongqing, Affiliated Hospital of Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Yuanmiao Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Jun Pan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Li Li
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Miao Wang
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Xiaoxue Ke
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Hongjuan Cui, ; Xiaoxue Ke,
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- *Correspondence: Hongjuan Cui, ; Xiaoxue Ke,
| |
Collapse
|
12
|
Ou J, Liu Q, Bian Y, Luan X, Meng Y, Dong H, Cao M, Zhang B, Wang Z, Zhao W. Integrated analysis of mRNA and microRNA transcriptome related to immunity and autophagy in shrimp hemocytes infected with Spiroplasma eriocheiris. FISH & SHELLFISH IMMUNOLOGY 2022; 130:436-452. [PMID: 36184970 DOI: 10.1016/j.fsi.2022.09.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
In recent years, the industry in charge of the cultivation of Macrobrachium nipponense (M.nipponense) has suffered significant economic losses due to an infectious pathogen called Spiroplasma eriocheiris (S.eriocheiris). There has therefore been a need to identify the key immune and autophagy genes that respond to M.nipponense's infection with S. eriocheiris to analyze its immune response mechanism and the regulation of related microRNAs (miRNAs). In this study, the mRNA and miRNA transcriptome of M.nipponense's hemocytes were analyzed at different stages of infection. This analysis employed the second and third-generation sequencing technologies. In the mRNA transcriptome, 1656 genes were expressed in healthy and susceptible M.nipponense. 892 of these were significantly up-regulated, while 764 were down-regulated. 118 genes with significant differences in autophagy, endocytosis, lysosome, Toll, IMD, and VEGF pathways were obtained from the transcriptome. In the miRNA transcriptome, 312 miRNAs (Conserved: 112, PN-type: 18, PC-type: 182) were sequenced. 74 were significantly up-regulated, and 57 were down-regulated. There were 25 miRNAs involved in regulating the Toll and IMD pathways, 41 in endocytosis, 30 in lysosome, and 12 in the VEGF pathway. An integrated analysis of immune-related miRNAs and mRNAs showed that miRNAs with significant differences (P < 0.05) such as ame-miR-29b-3p, dpu-miR-1and PC-3p-945_4074, had corresponding regulatory relationships with 118 important immune genes such as Relish, Dorsal, Caspase-3, and NF-κB. This study obtained the key immune and autophagy-related genes and corresponding regulatory miRNAs in M. nipponense's hemocytes in response to an infection by S.eriocheiris. The results can provide vital data that further reveals the defense mechanism of M.nipponense's immune system against S.eriocheiris. It can also help further comprehension and interpretation of M.nipponense's resistance mechanism to the invading S.eriocheiris, and provide molecular research information for the realization of host-directed therapies (HDT) for M.nipponense.
Collapse
Affiliation(s)
- Jiangtao Ou
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China.
| | - Qiao Liu
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China; The Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, Province Jiangsu, China
| | - Yunxia Bian
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Xiaoqi Luan
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China; Jiangsu Key Laboratory for Biodiversity & Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, China
| | - Yusuo Meng
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Huizi Dong
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Miao Cao
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Benhou Zhang
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Zisheng Wang
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| | - Weihong Zhao
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, School of Marine and Biological Engineering, Yancheng Institute of Technology, Yancheng, 224051, Province Jiangsu, China
| |
Collapse
|
13
|
Kim JK, Silwal P, Jo EK. Sirtuin 1 in Host Defense during Infection. Cells 2022; 11:cells11182921. [PMID: 36139497 PMCID: PMC9496836 DOI: 10.3390/cells11182921] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Sirtuins (SIRTs) are members of the class III histone deacetylase family and epigenetically control multiple target genes to modulate diverse biological responses in cells. Among the SIRTs, SIRT1 is the most well-studied, with a role in the modulation of immune and inflammatory responses following infection. The functions of SIRT1 include orchestrating immune, inflammatory, metabolic, and autophagic responses, all of which are required in establishing and controlling host defenses during infection. In this review, we summarize recent information on the roles of SIRT1 and its regulatory mechanisms during bacterial, viral, and parasitic infections. We also discuss several SIRT1 modulators, as potential antimicrobial treatments. Understanding the function of SIRT1 in balancing immune homeostasis will contribute to the development of new therapeutics for the treatment of infection and inflammatory disease.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu 42601, Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015, Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015, Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015, Korea
- Correspondence:
| |
Collapse
|
14
|
Lu G, Wang Y, Shi Y, Zhang Z, Huang C, He W, Wang C, Shen H. Autophagy in health and disease: From molecular mechanisms to therapeutic target. MedComm (Beijing) 2022; 3:e150. [PMID: 35845350 PMCID: PMC9271889 DOI: 10.1002/mco2.150] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionally conserved catabolic process in which cytosolic contents, such as aggregated proteins, dysfunctional organelle, or invading pathogens, are sequestered by the double-membrane structure termed autophagosome and delivered to lysosome for degradation. Over the past two decades, autophagy has been extensively studied, from the molecular mechanisms, biological functions, implications in various human diseases, to development of autophagy-related therapeutics. This review will focus on the latest development of autophagy research, covering molecular mechanisms in control of autophagosome biogenesis and autophagosome-lysosome fusion, and the upstream regulatory pathways including the AMPK and MTORC1 pathways. We will also provide a systematic discussion on the implication of autophagy in various human diseases, including cancer, neurodegenerative disorders (Alzheimer disease, Parkinson disease, Huntington's disease, and Amyotrophic lateral sclerosis), metabolic diseases (obesity and diabetes), viral infection especially SARS-Cov-2 and COVID-19, cardiovascular diseases (cardiac ischemia/reperfusion and cardiomyopathy), and aging. Finally, we will also summarize the development of pharmacological agents that have therapeutic potential for clinical applications via targeting the autophagy pathway. It is believed that decades of hard work on autophagy research is eventually to bring real and tangible benefits for improvement of human health and control of human diseases.
Collapse
Affiliation(s)
- Guang Lu
- Department of Physiology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yu Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yin Shi
- Department of BiochemistryZhejiang University School of MedicineHangzhouChina
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn ResearchSouthwest HospitalArmy Medical UniversityChongqingChina
| | - Chuang Wang
- Department of Pharmacology, Provincial Key Laboratory of PathophysiologyNingbo University School of MedicineNingboZhejiangChina
| | - Han‐Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision OncologyUniversity of MacauMacauChina
| |
Collapse
|
15
|
Kim JK, Park EJ, Jo EK. Itaconate, Arginine, and Gamma-Aminobutyric Acid: A Host Metabolite Triad Protective Against Mycobacterial Infection. Front Immunol 2022; 13:832015. [PMID: 35185924 PMCID: PMC8855927 DOI: 10.3389/fimmu.2022.832015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/13/2022] [Indexed: 12/29/2022] Open
Abstract
Immune metabolic regulation shapes the host-pathogen interaction during infection with Mycobacterium tuberculosis (Mtb), the pathogen of human tuberculosis (TB). Several immunometabolites generated by metabolic remodeling in macrophages are implicated in innate immune protection against Mtb infection by fine-tuning defensive pathways. Itaconate, produced by the mitochondrial enzyme immunoresponsive gene 1 (IRG1), has antimicrobial and anti-inflammatory effects, restricting intracellular mycobacterial growth. L-arginine, a component of the urea cycle, is critical for the synthesis of nitric oxide (NO) and is implicated in M1-mediated antimycobacterial responses in myeloid cells. L-citrulline, a by-product of NO production, contributes to host defense and generates L-arginine in myeloid cells. In arginase 1-expressing cells, L-arginine can be converted into ornithine, a polyamine precursor that enhances autophagy and antimicrobial protection against Mtb in Kupffer cells. Gamma-aminobutyric acid (GABA), a metabolite and neurotransmitter, activate autophagy to induce antimycobacterial host defenses. This review discusses the recent updates of the functions of the three metabolites in host protection against mycobacterial infection. Understanding the mechanisms by which these metabolites promote host defense will facilitate the development of novel host-directed therapeutics against Mtb and drug-resistant bacteria.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Eun-Jin Park
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, South Korea
- *Correspondence: Eun-Kyeong Jo,
| |
Collapse
|
16
|
Wu Y, Lin X, Song F, Xue D, Wang Y. Vitamin D3 promotes autophagy in THP‑1 cells infected with Mycobacterium tuberculosis. Exp Ther Med 2022; 23:240. [PMID: 35222717 PMCID: PMC8815057 DOI: 10.3892/etm.2022.11165] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/16/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yiming Wu
- Key Laboratory of The Ministry of Education for Conservation and Utilization of Special Biological Resources in The West, Yinchuan, Ningxia 750021, P.R. China
| | - Xue Lin
- Key Laboratory of The Ministry of Education for Conservation and Utilization of Special Biological Resources in The West, Yinchuan, Ningxia 750021, P.R. China
| | - Fuyang Song
- Key Laboratory of The Ministry of Education for Conservation and Utilization of Special Biological Resources in The West, Yinchuan, Ningxia 750021, P.R. China
| | - Di Xue
- Key Laboratory of The Ministry of Education for Conservation and Utilization of Special Biological Resources in The West, Yinchuan, Ningxia 750021, P.R. China
| | - Yujiong Wang
- Key Laboratory of The Ministry of Education for Conservation and Utilization of Special Biological Resources in The West, Yinchuan, Ningxia 750021, P.R. China
| |
Collapse
|
17
|
Liu H, Gui X, Chen S, Fu W, Li X, Xiao T, Hou J, Jiang T. Structural Variability of Lipoarabinomannan Modulates Innate Immune Responses within Infected Alveolar Epithelial Cells. Cells 2022; 11:cells11030361. [PMID: 35159170 PMCID: PMC8834380 DOI: 10.3390/cells11030361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
Mycobacterium tuberculosis (M. tb) is an intracellular pathogen persisting in phagosomes that has the ability to escape host immune surveillance causing tuberculosis (TB). Lipoarabinomannan (LAM), as a glycolipid, is one of the complex outermost components of the mycobacterial cell envelope and plays a critical role in modulating host responses during M. tb infection. Different species within the Mycobacterium genus exhibit distinct LAM structures and elicit diverse innate immune responses. However, little is known about the mechanisms. In this study, we first constructed a LAM-truncated mutant with fewer arabinofuranose (Araf) residues named M. sm-ΔM_6387 (Mycobacterium smegmatis arabinosyltransferase EmbC gene knockout strain). It exhibited some prominent cell wall defects, including tardiness of mycobacterial migration, loss of acid-fast staining, and increased cell wall permeability. Within alveolar epithelial cells (A549) infected by M. sm-ΔM_6387, the uptake rate was lower, phagosomes with bacterial degradation appeared, and microtubule-associated protein light chain 3 (LC3) recruitment was enhanced compared to wild type Mycobacterium smegmatis (M. smegmatis). We further confirmed that the variability in the removal capability of M. sm-ΔM_6387 resulted from host cell responses rather than the changes in the mycobacterial cell envelope. Moreover, we found that M. sm-ΔM_6387 or its glycolipid extracts significantly induced expression changes in some genes related to innate immune responses, including Toll-like receptor 2 (TLR2), class A scavenger receptor (SR-A), Rubicon, LC3, tumor necrosis factor alpha (TNF-α), Bcl-2, and Bax. Therefore, our studies suggest that nonpathogenic M. smegmatis can deposit LC3 on phagosomal membranes, and the decrease in the quantity of Araf residues for LAM molecules not only impacts mycobacterial cell wall integrity but also enhances host defense responses against the intracellular pathogens and decreases phagocytosis of host cells.
Collapse
Affiliation(s)
- Hanrui Liu
- Department of Biotechnology, The College of Basic Medical Science, Dalian Medical University, Dalian 116044, China; (H.L.); (X.G.); (W.F.); (X.L.); (T.X.); (J.H.)
| | - Xuwen Gui
- Department of Biotechnology, The College of Basic Medical Science, Dalian Medical University, Dalian 116044, China; (H.L.); (X.G.); (W.F.); (X.L.); (T.X.); (J.H.)
| | - Shixing Chen
- Key Laboratory of Science and Technology on Microsystem, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China;
| | - Weizhe Fu
- Department of Biotechnology, The College of Basic Medical Science, Dalian Medical University, Dalian 116044, China; (H.L.); (X.G.); (W.F.); (X.L.); (T.X.); (J.H.)
| | - Xiang Li
- Department of Biotechnology, The College of Basic Medical Science, Dalian Medical University, Dalian 116044, China; (H.L.); (X.G.); (W.F.); (X.L.); (T.X.); (J.H.)
| | - Tingyuan Xiao
- Department of Biotechnology, The College of Basic Medical Science, Dalian Medical University, Dalian 116044, China; (H.L.); (X.G.); (W.F.); (X.L.); (T.X.); (J.H.)
| | - Jie Hou
- Department of Biotechnology, The College of Basic Medical Science, Dalian Medical University, Dalian 116044, China; (H.L.); (X.G.); (W.F.); (X.L.); (T.X.); (J.H.)
| | - Tao Jiang
- Department of Biotechnology, The College of Basic Medical Science, Dalian Medical University, Dalian 116044, China; (H.L.); (X.G.); (W.F.); (X.L.); (T.X.); (J.H.)
- Correspondence: ; Tel.: +86-411-8611-0350
| |
Collapse
|
18
|
Blanco R, Gómez de Cedrón M, Gámez-Reche L, Martín-Leal A, González-Martín A, Lacalle RA, Ramírez de Molina A, Mañes S. The Chemokine Receptor CCR5 Links Memory CD4 + T Cell Metabolism to T Cell Antigen Receptor Nanoclustering. Front Immunol 2021; 12:722320. [PMID: 34950130 PMCID: PMC8688711 DOI: 10.3389/fimmu.2021.722320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
The inhibition of anabolic pathways, such as aerobic glycolysis, is a metabolic cornerstone of memory T cell differentiation and function. However, the signals that hamper these anabolic pathways are not completely known. Recent evidence pinpoints the chemokine receptor CCR5 as an important player in CD4+ T cell memory responses by regulating T cell antigen receptor (TCR) nanoclustering in an antigen-independent manner. This paper reports that CCR5 specifically restrains aerobic glycolysis in memory-like CD4+ T cells, but not in effector CD4+ T cells. CCR5-deficient memory CD4+ T cells thus show an abnormally high glycolytic/oxidative metabolism ratio. No CCR5-dependent change in glucose uptake nor in the expression of the main glucose transporters was detected in any of the examined cell types, although CCR5-deficient memory cells did show increased expression of the hexokinase 2 and pyruvate kinase M2 isoforms, plus the concomitant downregulation of Bcl-6, a transcriptional repressor of these key glycolytic enzymes. Further, the TCR nanoclustering defects observed in CCR5-deficient antigen-experienced CD4+ T cells were partially reversed by incubation with 2-deoxyglucose (2-DG), suggesting a link between inhibition of the glycolytic pathway and TCR nanoscopic organization. Indeed, the treatment of CCR5-deficient lymphoblasts with 2-DG enhanced IL-2 production after antigen re-stimulation. These results identify CCR5 as an important regulator of the metabolic fitness of memory CD4+ T cells, and reveal an unexpected link between T cell metabolism and TCR organization with potential influence on the response of memory T cells upon antigen re-encounter.
Collapse
Affiliation(s)
- Raquel Blanco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB/CSIC), Madrid, Spain
| | - Marta Gómez de Cedrón
- Precision Nutrition and Cancer Program, Molecular Oncology Group, IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| | - Laura Gámez-Reche
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB/CSIC), Madrid, Spain.,Department of Biochemistry, Universidad Autónoma de Madrid, and Instituto de Investigaciones Biomédicas Alberto Sols (IIB/CSIC), Madrid, Spain
| | - Ana Martín-Leal
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB/CSIC), Madrid, Spain
| | - Alicia González-Martín
- Department of Biochemistry, Universidad Autónoma de Madrid, and Instituto de Investigaciones Biomédicas Alberto Sols (IIB/CSIC), Madrid, Spain
| | - Rosa A Lacalle
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB/CSIC), Madrid, Spain
| | - Ana Ramírez de Molina
- Precision Nutrition and Cancer Program, Molecular Oncology Group, IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB/CSIC), Madrid, Spain
| |
Collapse
|
19
|
Zhang W, Xu L, Zhu L, Liu Y, Yang S, Zhao M. Lipid Droplets, the Central Hub Integrating Cell Metabolism and the Immune System. Front Physiol 2021; 12:746749. [PMID: 34925055 PMCID: PMC8678573 DOI: 10.3389/fphys.2021.746749] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Lipid droplets (LDs) are commonly found in various biological cells and are organelles related to cell metabolism. LDs, the number and size of which are heterogeneous across cell type, are primarily composed of polar lipids and proteins on the surface with neutral lipids in the core. Neutral lipids stored in LDs can be degraded by lipolysis and lipophagocytosis, which are regulated by various proteins. The process of LD formation can be summarized in four steps. In addition to energy production, LDs play an extremely pivotal role in a variety of physiological and pathological processes, such as endoplasmic reticulum stress, lipid toxicity, storage of fat-soluble vitamins, regulation of oxidative stress, and reprogramming of cell metabolism. Interestingly, LDs, the hub of integration between metabolism and the immune system, are involved in antitumor immunity, anti-infective immunity (viruses, bacteria, parasites, etc.) and some metabolic immune diseases. Herein, we summarize the role of LDs in several major immune cells as elucidated in recent years, including T cells, dendritic cells, macrophages, mast cells, and neutrophils. Additionally, we analyze the role of the interaction between LDs and immune cells in two typical metabolic immune diseases: atherosclerosis and Mycobacterium tuberculosis infection.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya Hospital, Central South University, Changsha, China
| | - Linyong Xu
- School of Life Sciences, Central South University, Changsha, China
| | - Ling Zhu
- School of Life Sciences, Central South University, Changsha, China
| | - Yifan Liu
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Siwei Yang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Mingyi Zhao
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Bittencourt TL, da Silva Prata RB, de Andrade Silva BJ, de Mattos Barbosa MG, Dalcolmo MP, Pinheiro RO. Autophagy as a Target for Drug Development Of Skin Infection Caused by Mycobacteria. Front Immunol 2021; 12:674241. [PMID: 34113346 PMCID: PMC8185338 DOI: 10.3389/fimmu.2021.674241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
Pathogenic mycobacteria species may subvert the innate immune mechanisms and can modulate the activation of cells that cause disease in the skin. Cutaneous mycobacterial infection may present different clinical presentations and it is associated with stigma, deformity, and disability. The understanding of the immunopathogenic mechanisms related to mycobacterial infection in human skin is of pivotal importance to identify targets for new therapeutic strategies. The occurrence of reactional episodes and relapse in leprosy patients, the emergence of resistant mycobacteria strains, and the absence of effective drugs to treat mycobacterial cutaneous infection increased the interest in the development of therapies based on repurposed drugs against mycobacteria. The mechanism of action of many of these therapies evaluated is linked to the activation of autophagy. Autophagy is an evolutionary conserved lysosomal degradation pathway that has been associated with the control of the mycobacterial bacillary load. Here, we review the role of autophagy in the pathogenesis of cutaneous mycobacterial infection and discuss the perspectives of autophagy as a target for drug development and repurposing against cutaneous mycobacterial infection.
Collapse
Affiliation(s)
| | | | | | | | - Margareth Pretti Dalcolmo
- Helio Fraga Reference Center, Sergio Arouca National School of Public Health, Fiocruz, Rio de Janeiro, Brazil
| | - Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Silwal P, Kim JK, Jeon SM, Lee JY, Kim YJ, Kim YS, Seo Y, Kim J, Kim SY, Lee MJ, Heo JY, Jung MJ, Kim HS, Hyun DW, Han JE, Whang J, Huh YH, Lee SH, Heo WD, Kim JM, Bae JW, Jo EK. Mitofusin-2 boosts innate immunity through the maintenance of aerobic glycolysis and activation of xenophagy in mice. Commun Biol 2021; 4:548. [PMID: 33972668 PMCID: PMC8110749 DOI: 10.1038/s42003-021-02073-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/29/2021] [Indexed: 02/08/2023] Open
Abstract
Mitochondrial function and innate immunity are intimately linked; however, the mechanisms how mitochondrion-shaping proteins regulate innate host defense remains largely unknown. Herein we show that mitofusin-2 (MFN2), a mitochondrial fusion protein, promotes innate host defense through the maintenance of aerobic glycolysis and xenophagy via hypoxia-inducible factor (HIF)-1α during intracellular bacterial infection. Myeloid-specific MFN2 deficiency in mice impaired the antimicrobial and inflammatory responses against mycobacterial and listerial infection. Mechanistically, MFN2 was required for the enhancement of inflammatory signaling through optimal induction of aerobic glycolysis via HIF-1α, which is activated by mitochondrial respiratory chain complex I and reactive oxygen species, in macrophages. MFN2 did not impact mitophagy during infection; however, it promoted xenophagy activation through HIF-1α. In addition, MFN2 interacted with the late endosomal protein Rab7, to facilitate xenophagy during mycobacterial infection. Our findings reveal the mechanistic regulations by which MFN2 tailors the innate host defense through coordinated control of immunometabolism and xenophagy via HIF-1α during bacterial infection.
Collapse
Affiliation(s)
- Prashanta Silwal
- grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jin Kyung Kim
- grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sang Min Jeon
- grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - June-Young Lee
- grid.289247.20000 0001 2171 7818Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Young Jae Kim
- grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Yi Sak Kim
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California, San Diego, CA USA
| | - Yeji Seo
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Jihye Kim
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Soo Yeon Kim
- grid.418980.c0000 0000 8749 5149Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Min Joung Lee
- grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jun Young Heo
- grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Korea
| | - Mi-Ja Jung
- grid.289247.20000 0001 2171 7818Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Hyun Sik Kim
- grid.289247.20000 0001 2171 7818Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Dong-Wook Hyun
- grid.289247.20000 0001 2171 7818Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Jeong Eun Han
- grid.289247.20000 0001 2171 7818Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC) & Basic Research Section, The Korean Institute of Tuberculosis (KIT), Cheongju, Korea
| | - Yang Hoon Huh
- grid.410885.00000 0000 9149 5707Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| | - Sang-Hee Lee
- grid.410885.00000 0000 9149 5707Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| | - Won Do Heo
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Jin-Man Kim
- grid.254230.20000 0001 0722 6377Department of Pathology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jin-Woo Bae
- grid.289247.20000 0001 2171 7818Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Eun-Kyeong Jo
- grid.254230.20000 0001 0722 6377Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea ,grid.254230.20000 0001 0722 6377Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
22
|
Etna MP, Severa M, Licursi V, Pardini M, Cruciani M, Rizzo F, Giacomini E, Macchia G, Palumbo O, Stallone R, Carella M, Livingstone M, Negri R, Pellegrini S, Coccia EM. Genome-Wide Gene Expression Analysis of Mtb-Infected DC Highlights the Rapamycin-Driven Modulation of Regulatory Cytokines via the mTOR/GSK-3β Axis. Front Immunol 2021; 12:649475. [PMID: 33936070 PMCID: PMC8086600 DOI: 10.3389/fimmu.2021.649475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
In human primary dendritic cells (DC) rapamycin-an autophagy inducer and protein synthesis inhibitor-overcomes the autophagy block induced by Mycobacterium tuberculosis (Mtb) and promotes a Th1 response via IL-12 secretion. Here, the immunostimulatory activity of rapamycin in Mtb-infected DC was further investigated by analyzing both transcriptome and translatome gene profiles. Hundreds of differentially expressed genes (DEGs) were identified by transcriptome and translatome analyses of Mtb-infected DC, and some of these genes were found further modulated by rapamycin. The majority of transcriptome-associated DEGs overlapped with those present in the translatome, suggesting that transcriptionally stimulated mRNAs are also actively translated. In silico analysis of DEGs revealed significant changes in intracellular cascades related to cytokine production, cytokine-induced signaling and immune response to pathogens. In particular, rapamycin treatment of Mtb-infected DC caused an enrichment of IFN-β, IFN-λ and IFN-stimulated gene transcripts in the polysome-associated RNA fraction. In addition, rapamycin led to an increase of IL-12, IL-23, IL-1β, IL-6, and TNF-α but to a reduction of IL-10. Interestingly, upon silencing or pharmacological inhibition of GSK-3β, the rapamycin-driven modulation of the pro- and anti-inflammatory cytokine balance was lost, indicating that, in Mtb-infected DC, GSK-3β acts as molecular switch for the regulation of the cytokine milieu. In conclusion, our study sheds light on the molecular mechanism by which autophagy induction contributes to DC activation during Mtb infection and points to rapamycin and GSK-3β modulators as promising compounds for host-directed therapy in the control of Mtb infection.
Collapse
Affiliation(s)
- Marilena P Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology, Sapienza University, Rome, Italy
| | - Manuela Pardini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Melania Cruciani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Raffaella Stallone
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Massimo Carella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Mark Livingstone
- Cytokine Signaling Unit, Inserm, Institut Pasteur, Paris, France
| | - Rodolfo Negri
- Department of Biology and Biotechnology, Sapienza University, Rome, Italy
| | | | - Eliana M Coccia
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|