1
|
Sun W, Wu H, Zhao G, Shui Q, Zhang L, Luan X, Chen T, Liu F, Zheng Y, Zhao W, Qi X, Liu B, Gao C. Lipid droplets restrict phagosome formation in antifungal immunity. Cell Mol Immunol 2025; 22:468-484. [PMID: 40195475 PMCID: PMC12041225 DOI: 10.1038/s41423-025-01282-x] [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: 09/18/2024] [Accepted: 03/14/2025] [Indexed: 04/09/2025] Open
Abstract
Lipid droplets (LDs) are intracellular organelles that can be induced and interact with phagosomes during the process of pathogen phagocytosis in macrophages. However, the function of LDs in phagocytosis remains elusive. Here, we unveil the role of LDs in modulating phagosome formation via a fungal infection model. Specifically, LD accumulation restricted the degree of phagosome formation and protected macrophages from death. Mechanistically, LD formation competitively consumed the intracellular endoplasmic reticulum membrane and altered RAC1 translocation and GTPase activity, which resulted in limited phagosome formation in macrophages during fungal engulfment. Mice with Hilpda-deficient macrophages were more susceptible to the lethal sequelae of systemic infection with C. albicans. Notably, administration of the ATGL inhibitor atglistatin improved host outcomes in disseminated fungal infections. Taken together, our study elucidates the mechanism by which LDs control phagosome formation to prevent immune cell death and offers a potential drug target for the treatment of C. albicans infections.
Collapse
Affiliation(s)
- Wanwei Sun
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Han Wu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Guimin Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Qing Shui
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Xiaoxi Luan
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Tian Chen
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Feng Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Xiaopeng Qi
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China.
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China.
| |
Collapse
|
2
|
Lösslein AK, Henneke P. Macrophage Differentiation and Metabolic Adaptation in Mycobacterial Infections. Annu Rev Immunol 2025; 43:423-450. [PMID: 40014665 DOI: 10.1146/annurev-immunol-082323-120757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
The adaptation of macrophages-the most common tissue-resident immune cells-to metabolic and microbial cues with high local variability is essential for the maintenance of organ integrity. In homeostasis, macrophages show largely predictable tissue-specific differentiation, as recently revealed by multidimensional methods. However, chronic infections with human-adapted pathogens substantially contribute to the differentiation complexity of tissue macrophages, which has been only partially resolved. Specifically, the response to mycobacterial species-which range from Mycobacterium tuberculosis (with highest specificity for humans, broad organ tropism, yet tissue-specific disease phenotypes) to environmental mycobacteria with humans as accidental hosts-may serve as a paradigm of tissue macrophage adaptation mechanisms. While mycobacterial species-specific tissue preferences are partially related to the mode of acquisition and pathogen characteristics, evolutionary convergence with macrophages driven by metabolic features of the target organ likely contributes to infection resistance and immunopathology. In this review, we unravel the mechanisms of tissue-specific macrophage differentiation and its limitations in mycobacterial infections.
Collapse
Affiliation(s)
- Anne Kathrin Lösslein
- Institute for Infection Prevention and Control, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany;
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Institute for Infection Prevention and Control, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany;
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
3
|
Valeriano JDP, Andrade-Silva M, Pereira-Dutra F, Seito LN, Bozza PT, Rosas EC, Souza Costa MF, Henriques MG. Cannabinoid receptor type 2 agonist GP1a attenuates macrophage activation induced by M. bovis-BCG by inhibiting NF-κB signaling. J Leukoc Biol 2025; 117:qiae246. [PMID: 39538989 DOI: 10.1093/jleuko/qiae246] [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: 07/18/2024] [Revised: 08/28/2024] [Accepted: 11/13/2024] [Indexed: 11/16/2024] Open
Abstract
Tuberculosis (TB) is one of the leading causes of death worldwide and a major public health problem. Immune evasion mechanisms and antibiotic resistance highlight the need to better understand this disease and explore alternative treatment approaches. Mycobacterial infection modulates the macrophage response and metabolism to persist and proliferate inside the cell. Cannabinoid receptor type 2 (CB2) is expressed mainly in leukocytes and modulates the course of inflammatory diseases. Therefore, our study aimed to evaluate the effects of the CB2-selective agonist GP1a on irradiated Mycobacterium bovis-BCG (iBCG)-induced J774A.1 macrophage activation. We observed increased expression of CB2 in macrophages after iBCG stimulation. The pretreatment with CB2-agonists, GP1a, JWH-133, and GW-833972A (10 µM), reduced iBCG-induced TNF-α and IL-6 release by these cells. Moreover, the CB2-antagonist AM630 (200 nM) treatment confirmed the activity of GP1a on CB2 by scale down its effect on cytokine production. GP1a pretreatment (10 µM) also inhibited the iBCG-induced production of inflammatory mediators as prostaglandin (PG)E2 and nitric oxide by macrophages. Additionally, GP1a pretreatment also reduced the transcription of proinflammatory genes (inos, il1b, and cox2) and genes related to lipid metabolism (dgat1, acat1, plin2, atgl, and cd36). Indeed, lipid droplet accumulation was reduced by GP1a treatment, which was partially blockade by AM630 pretreatment. Finally, GP1a pretreatment reduced the activation of the NF-κB signaling pathway. In conclusion, the activation of CB2 by GP1a modulated the macrophage response to iBCG by reducing inflammatory mediator levels and metabolic reprogramming.
Collapse
Affiliation(s)
- Jessica Do Prado Valeriano
- Immunobiology Department, Immunobiology of Inflammation Laboratory, IB, Universidade Federal Fluminense, R. Prof. Marcos Waldemar de Freitas Reis - São Domingos, Niterói - RJ 24210-201, Brazil
- Graduate Program in Biosciences-IBRAG IBRAG, Universidade do Estado do Rio de Janeiro, Blvd. 28 de Setembro, 87 - fundos - Vila Isabel, Rio de Janeiro - RJ 20551-030, Brazil
| | - Magaiver Andrade-Silva
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rua Sizenando Nabuco, 100, Manguinhos, Rio de Janeiro - RJ 21041-000, Brazil
| | - Filipe Pereira-Dutra
- Immunopharmacology Laboratory, IOC, Oswaldo Cruz Foundation, Av. Brasil, 4365 - Manguinhos, Rio de Janeiro - RJ 21040-900, Brazil
| | - Leonardo Noboru Seito
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rua Sizenando Nabuco, 100, Manguinhos, Rio de Janeiro - RJ 21041-000, Brazil
| | - Patricia Torres Bozza
- Immunopharmacology Laboratory, IOC, Oswaldo Cruz Foundation, Av. Brasil, 4365 - Manguinhos, Rio de Janeiro - RJ 21040-900, Brazil
| | - Elaine Cruz Rosas
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rua Sizenando Nabuco, 100, Manguinhos, Rio de Janeiro - RJ 21041-000, Brazil
| | - Maria Fernanda Souza Costa
- Immunobiology Department, Immunobiology of Inflammation Laboratory, IB, Universidade Federal Fluminense, R. Prof. Marcos Waldemar de Freitas Reis - São Domingos, Niterói - RJ 24210-201, Brazil
| | - Maria G Henriques
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rua Sizenando Nabuco, 100, Manguinhos, Rio de Janeiro - RJ 21041-000, Brazil
| |
Collapse
|
4
|
Simwela NV, Jaecklein E, Sassetti CM, Russell DG. Impaired fatty acid import or catabolism in macrophages restricts intracellular growth of Mycobacterium tuberculosis. eLife 2025; 13:RP102980. [PMID: 40080408 PMCID: PMC11906158 DOI: 10.7554/elife.102980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025] Open
Abstract
Mycobacterium tuberculosis (Mtb) infection of macrophages reprograms cellular metabolism to promote lipid retention. While it is clearly known that intracellular Mtb utilize host-derived lipids to maintain infection, the role of macrophage lipid processing on the bacteria's ability to access the intracellular lipid pool remains undefined. We utilized a CRISPR-Cas9 genetic approach to assess the impact of sequential steps in fatty acid metabolism on the growth of intracellular Mtb. Our analyses demonstrate that macrophages that cannot either import, store, or catabolize fatty acids restrict Mtb growth by both common and divergent antimicrobial mechanisms, including increased glycolysis, increased oxidative stress, production of pro-inflammatory cytokines, enhanced autophagy, and nutrient limitation. We also show that impaired macrophage lipid droplet biogenesis is restrictive to Mtb replication, but increased induction of the same fails to rescue Mtb growth. Our work expands our understanding of how host fatty acid homeostasis impacts Mtb growth in the macrophage.
Collapse
Affiliation(s)
- Nelson V Simwela
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Eleni Jaecklein
- Department of Microbiology, UMass Chan Medical SchoolWorcesterUnited States
| | | | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| |
Collapse
|
5
|
Peng Y, Liu Y, Wu J, Zhang Y, Wang Q, Shao S. ArgR-dependent bacterial resistance to host lipid droplets in Edwardsiella piscicida. Commun Biol 2025; 8:333. [PMID: 40021749 PMCID: PMC11871307 DOI: 10.1038/s42003-025-07777-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Lipid droplets (LDs), as innate immune hubs, function in the front line of antimicrobial defense involved in the host-pathogen arms race. Particularly for intracellular bacterial pathogens, the endowed capacity to resist host LDs can effectively facilitate pathogen in vivo colonization and evasion from the host's innate immune response. Here, to investigate the genetic mechanisms of intracellular bacteria response to host LDs, we utilized transposon insertion sequencing to dissect critical fitness determinants of Edwardsiella piscicida under the treatment of LDs isolated from its native host, turbot. Targeted metabolomics indicated that LD challenge resulted in the accumulation of intracellular arginine. The core arginine metabolism regulatory factor, ArgR, was found to play a pivotal role in combating LDs, emphasizing the importance of orchestrating intracellular arginine levels for bacterial LD adaptation. Specifically, ArgR enhanced the expressions of genes involved in arginine catabolism (speA/B and arcC) and diminished gene transcripts associated with arginine import (artP) and synthesis (argD/E/H). Furthermore, ArgR contributed to the pathogenesis of E. piscicida, promoting the proliferation in host cells and virulence in turbot. Collectively, our results shed light on the underlying mechanism of intracellular pathogens resisting LDs during bacterial infections and highlighting the crucial role of arginine in the host-pathogen interactions.
Collapse
Affiliation(s)
- Yue Peng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yihan Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Junze Wu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuanxing Zhang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), 519000, Zhuhai, China
| | - Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, China
- Laboratory of Aquatic Animal Diseases of MOA, Shanghai, China
| | - Shuai Shao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, China.
- Laboratory of Aquatic Animal Diseases of MOA, Shanghai, China.
| |
Collapse
|
6
|
Oleszycka E, Kwiecień K, Grygier B, Cichy J, Kwiecińska P. The many faces of DGAT1. Life Sci 2025; 362:123322. [PMID: 39709166 DOI: 10.1016/j.lfs.2024.123322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) is a multifaced enzyme with a wide spectrum of substrates, from lipids through waxes to retinoids, which makes it an interesting therapeutic target. DGAT1 inhibitors are currently at various stages of preclinical and clinical trials, mostly related to metabolic diseases. Interestingly, in recent years, a growing amount of research has shown the influence of DGAT1 on immune cell metabolism and functions, highlighting its important role during infections and tumorigenesis. In this review, we aim to elucidate the potential immunomodulatory effect of DGAT1 in physiological and pathological conditions.
Collapse
Affiliation(s)
- Ewa Oleszycka
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Kamila Kwiecień
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Beata Grygier
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Science, Cracow, Poland
| | - Joanna Cichy
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Patrycja Kwiecińska
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland; Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland.
| |
Collapse
|
7
|
Simwela NV, Jaecklein E, Sassetti CM, Russell DG. Impaired fatty acid import or catabolism in macrophages restricts intracellular growth of Mycobacterium tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.22.604660. [PMID: 39091727 PMCID: PMC11291043 DOI: 10.1101/2024.07.22.604660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Mycobacterium tuberculosis (Mtb) infection of macrophages reprograms cellular metabolism to promote lipid retention. While it is clearly known that intracellular Mtb utilize host derived lipids to maintain infection, the role of macrophage lipid processing on the bacteria's ability to access the intracellular lipid pool remains undefined. We utilized a CRISPR-Cas9 genetic approach to assess the impact of sequential steps in fatty acid metabolism on the growth of intracellular Mtb. Our analyzes demonstrate that macrophages which cannot either import, store or catabolize fatty acids restrict Mtb growth by both common and divergent anti-microbial mechanisms, including increased glycolysis, increased oxidative stress, production of pro-inflammatory cytokines, enhanced autophagy and nutrient limitation. We also show that impaired macrophage lipid droplet biogenesis is restrictive to Mtb replication, but increased induction of the same fails to rescue Mtb growth. Our work expands our understanding of how host fatty acid homeostasis impacts Mtb growth in the macrophage.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Eleni Jaecklein
- Department of Microbiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | | | - David G. Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
8
|
Alvarez HM, Lanfranconi MP, Hernández MA. Metabolism-lipid droplet-nucleic acid crosstalk to regulate lipid storage and other cellular processes in oleaginous Rhodococcus bacteria. Biol Cell 2025; 117:e2400094. [PMID: 39853774 DOI: 10.1111/boc.202400094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/14/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025]
Abstract
Actinobacteria belonging to Mycobacterium and Rhodococcus genera are able to synthesize and intracellularly accumulate variable amounts of triacylglycerols (TAG) in the form of lipid droplets (LDs). The lipid storage capacity of LDs in cells is controlled by the balance between lipogenesis and lipolysis. The growth of LDs in bacterial cells may be directly promoted by TAG biosynthesis, whereas TAG degradation might result in the reduction of LD sizes and lipid storage capacity. Therefore, LD formation and turnover have to be precisely regulated to maintain a balanced lipid distribution, coupling gene regulation with the metabolic state of the cell. In eukaryotic cells, LDs have emerged as critical mediators of diverse cellular responses, including fatty acid trafficking and modulation of transcriptional programs. Recent studies performed in mycobacteria and rhodococci suggested the existence of similar crosstalk mechanisms between lipid metabolism, LDs, and gene expression regulation in cells. This review connects and organizes results of different studies in a comprehensive framework for providing evidence of "lipid metabolism-LDs-genomic DNA" crosstalk occurring in TAG-accumulating actinobacteria. We provide examples indicating that bacterial cells evolved sensing mechanisms that detect lipid metabolites changes as indicators of metabolic states, and adapt their transcriptional profiles through epigenetic-like mechanisms mediated by LD-associated proteins. Here, we describe the molecular interconnections of this coupling system and the main role of each component that integrates the information about the cellular metabolic state into the regulation of lipogenesis, LD formation and transcription in oleaginous bacteria.
Collapse
Affiliation(s)
- Héctor M Alvarez
- INBIOP (Instituto de Biociencias de la Patagonia), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Naturales y Ciencias de la Salud, Universidad Nacional de la Patagonia San Juan Bosco, Comodoro Rivadavia, Chubut, Argentina
| | - Mariana P Lanfranconi
- INBIOP (Instituto de Biociencias de la Patagonia), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Naturales y Ciencias de la Salud, Universidad Nacional de la Patagonia San Juan Bosco, Comodoro Rivadavia, Chubut, Argentina
| | - Martín A Hernández
- INBIOP (Instituto de Biociencias de la Patagonia), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Naturales y Ciencias de la Salud, Universidad Nacional de la Patagonia San Juan Bosco, Comodoro Rivadavia, Chubut, Argentina
| |
Collapse
|
9
|
Kim H, Kim SJ. 3D Bioprinting of Pig Macrophages and Human Cells Discovered the P2Y14 Receptor as a Mediator of Xenogenic Immune Responses. Immunol Invest 2025; 54:18-33. [PMID: 39356134 DOI: 10.1080/08820139.2024.2411388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
BACKGROUND The survival rate of pig lung xenotransplantation (PLXTx) recipients is severely limited by intense xenogenic immune responses, necessitating further insights into xenogeneic immunity and the development of models to study the PLXTx immune response. METHODS We identified regulators of PLXTx immune response Using Gene ontology analysis. We assessed the metabolic changes and protein levels in 3D4/31 pig alveolar macrophages (PAMs) through flow cytometry and immunoblotting. To induce a xenogenic immune response, we co-cultured 3D4/31-PAMs with A549 human alveolar epithelial cells and evaluated cytokine expression using qRT-PCR. RESULTS Gene ontology analysis identified STAT1 and alveolar macrophages as contributors to lung autoimmunity and transplant rejection. In 3D4/31-PAMs, phorbol myristate acetate-induced glycogen accumulation and cyclooxygenase-2 expression were inhibited by the P2Y14 inhibitor PPTN. Co-culturing 3D4/31-PAMs with A549 human alveolar epithelial cells via 3D bioprinting resulted in a more pronounced inflammatory response than 2D co-culture, with increased expression of genes related to the P2Y14 cascade and inflammation. This inflammatory gene expression was prevented by PPTN treatment. CONCLUSION Based on these results, we propose alginate bioprinting as an in vitro model for PLXTx and suggest that P2Y14 is a key regulator of xenogeneic immune responses in PAMs.
Collapse
Affiliation(s)
- Hyungkuen Kim
- Department of Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, Republic of Korea
| | - Sung-Jo Kim
- Department of Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, Republic of Korea
| |
Collapse
|
10
|
Magana-Arachchi D, Madegedara D, Bandara U. Whole Blood vs Serum-Derived Exosomes for Host and Pathogen-Specific Tuberculosis Biomarker Identification: RNA-Seq-Based Machine-Learning Approach. Biochem Genet 2024:10.1007/s10528-024-11002-1. [PMID: 39715973 DOI: 10.1007/s10528-024-11002-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Mycobacterium tuberculosis (Mtb) remains a leading infectious disease responsible for millions of deaths. RNA sequencing is a rapidly growing technique and a powerful approach to understanding host and pathogen cross-talks via transcriptional responses. However, its application is limited due to the high costs involved.This study is a preliminary attempt to understand host-pathogen cross-talk during TB infection in different TB clinical cohorts using two biological fluids: Whole blood and serum exosomes (EXO). We conducted an RNA-sequencing machine-learning approach using 20 active TB (ATB), 11 latent TB (LTB), three healthy control (HC) whole blood datasets, and two ATB, LTB, and HC serum EXO datasets. During the study, host-derived differentially expressed genes (DEGs) were identified in both whole blood and EXOs, while EXOs were successful in identifying pathogen-derived DEGs only in LTB. The majority of the DEGs in whole blood were up-regulated between ATB and HC, and ATB and LTB, while down-regulated between LTB and HC, which was vice versa for the EXOs, indicating different mechanisms in response to different states of TB infection across the two different biological samples. The pathway analysis revealed that whole blood gene signatures were mainly involved in host immune responses, whereas exosomal gene signatures were involved in manipulating the host's cellular responses and supporting Mtb survival. Overall, identifying both host and pathogen-derived gene signatures in different biological samples for intracellular pathogens like Mtb is vital to decipher the complex interplay between the host and the pathogen, ultimately leading to more successful future interventions.
Collapse
Affiliation(s)
| | | | - Upeka Bandara
- National Institute of Fundamental Studies, Kandy, Sri Lanka
| |
Collapse
|
11
|
Fu S, Yu R, Yang B, Han X, Xu Y, Miao J. Hypoxia-inducible lipid droplet-associated protein (HILPDA) and cystathionine β-synthase (CBS) co-contribute to protecting intestinal epithelial cells from Staphylococcus aureus via regulating lipid droplets formation. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159558. [PMID: 39173873 DOI: 10.1016/j.bbalip.2024.159558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Despite Staphylococcus aureus (S. aureus) being a highly studied zoontic bacterium, its enteropathogenicity remains elusive. Herein, our findings demonstrated that S. aureus infection led to the accumulation of lipid droplets (LDs) in intestinal epithelial cells, accompanied by marked elevation inflammatory response that ultimately decreases intracellular bacterial load. The aforestated phenomenon may be partly attributed to the up-regulation of hypoxia-inducible lipid droplet-associated protein (HILPDA) and the concomitant down-regulation of cystathionine β-synthase (CBS) protein. Moreover, S. aureus infection up-regulated the expression of HILPDA, thereby promoting LDs accumulation, and down-regulated that of CBS, consequently inhibiting microsomal triglyceride transfer protein (MTTP) expression. This process may suppress the transport of LDs to the extracellular environment, further contributing to the formation of intracellular LDs. In summary, the results of this study provide significant insights into the intricate mechanisms through which the host organism combats pathogens and maintains the balance of sulfur and lipid metabolism. These findings not only enhance our understanding of the host's defense mechanisms but also offer promising avenues for the development of novel strategies to combat intestinal infectious diseases.
Collapse
Affiliation(s)
- Shaodong Fu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Rui Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Bo Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
12
|
Andrews JT, Zhang Z, Prasad GVRK, Huey F, Nazarova EV, Wang J, Ranaraja A, Weinkopff T, Li LX, Mu S, Birrer MJ, Huang SCC, Zhang N, Argüello RJ, Philips JA, Mattila JT, Huang L. Metabolically active neutrophils represent a permissive niche for Mycobacterium tuberculosis. Mucosal Immunol 2024; 17:825-842. [PMID: 38844208 PMCID: PMC11493682 DOI: 10.1016/j.mucimm.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Mycobacterium tuberculosis (Mtb)-infected neutrophils are often found in the airways of patients with active tuberculosis (TB), and excessive recruitment of neutrophils to the lung is linked to increased bacterial burden and aggravated pathology in TB. The basis for the permissiveness of neutrophils for Mtb and the ability to be pathogenic in TB has been elusive. Here, we identified metabolic and functional features of neutrophils that contribute to their permissiveness in Mtb infection. Using single-cell metabolic and transcriptional analyses, we found that neutrophils in the Mtb-infected lung displayed elevated mitochondrial metabolism, which was largely attributed to the induction of activated neutrophils with enhanced metabolic activities. The activated neutrophil subpopulation was also identified in the lung granulomas from Mtb-infected non-human primates. Functionally, activated neutrophils harbored more viable bacteria and displayed enhanced lipid uptake and accumulation. Surprisingly, we found that interferon-γ promoted the activation of lung neutrophils during Mtb infection. Lastly, perturbation of lipid uptake pathways selectively compromised Mtb survival in activated neutrophils. These findings suggest that neutrophil heterogeneity and metabolic diversity are key to their permissiveness for Mtb and that metabolic pathways in neutrophils represent potential host-directed therapeutics in TB.
Collapse
Affiliation(s)
- J Tucker Andrews
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Zijing Zhang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - G V R Krishna Prasad
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Fischer Huey
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Evgeniya V Nazarova
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Jocelyn Wang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Ananya Ranaraja
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lin-Xi Li
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Shengyu Mu
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Michael J Birrer
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Stanley Ching-Cheng Huang
- Pelotonia Institute for Immuno-Oncology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Nan Zhang
- Immunology, Metastasis & Microenvironment Program, Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA, USA
| | - Rafael J Argüello
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Jennifer A Philips
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA; Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lu Huang
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
13
|
Roe K. Are secondary bacterial pneumonia mortalities increased because of insufficient pro-resolving mediators? J Infect Chemother 2024; 30:959-970. [PMID: 38977072 DOI: 10.1016/j.jiac.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/24/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
Respiratory viral infections, including respiratory syncytial virus (RSV), parainfluenza viruses and type A and B influenza viruses, can have severe outcomes. Bacterial infections frequently follow viral infections, and influenza or other viral epidemics periodically have higher mortalities from secondary bacterial pneumonias. Most secondary bacterial infections can cause lung immunosuppression by fatty acid mediators which activate cellular receptors to manipulate neutrophils, macrophages, natural killer cells, dendritic cells and other lung immune cells. Bacterial infections induce synthesis of inflammatory mediators including prostaglandins and leukotrienes, then eventually also special pro-resolving mediators, including lipoxins, resolvins, protectins and maresins, which normally resolve inflammation and immunosuppression. Concurrent viral and secondary bacterial infections are more dangerous, because viral infections can cause inflammation and immunosuppression before the secondary bacterial infections worsen inflammation and immunosuppression. Plausibly, the higher mortalities of secondary bacterial pneumonias are caused by the overwhelming inflammation and immunosuppression, which the special pro-resolving mediators might not resolve.
Collapse
Affiliation(s)
- Kevin Roe
- Retired United States Patent and Trademark Office, San Jose, CA, USA.
| |
Collapse
|
14
|
Bussi C, Lai R, Athanasiadi N, Gutierrez MG. Physiologic medium renders human iPSC-derived macrophages permissive for M. tuberculosis by rewiring organelle function and metabolism. mBio 2024; 15:e0035324. [PMID: 38984828 PMCID: PMC11323749 DOI: 10.1128/mbio.00353-24] [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: 02/05/2024] [Accepted: 06/04/2024] [Indexed: 07/11/2024] Open
Abstract
In vitro studies are crucial for our understanding of the human macrophage immune functions. However, traditional in vitro culture media poorly reflect the metabolic composition of blood, potentially affecting the outcomes of these studies. Here, we analyzed the impact of a physiological medium on human induced pluripotent stem cell (iPSC)-derived macrophages (iPSDM) function. Macrophages cultured in a human plasma-like medium (HPLM) were more permissive to Mycobacterium tuberculosis (Mtb) replication and showed decreased lipid metabolism with increased metabolic polarization. Functionally, we discovered that HPLM-differentiated macrophages showed different metabolic organelle content and activity. Specifically, HPLM-differentiated macrophages displayed reduced lipid droplet and peroxisome content, increased lysosomal proteolytic activity, and increased mitochondrial activity and dynamics. Inhibiting or inducing lipid droplet formation revealed that lipid droplet content is a key factor influencing macrophage permissiveness to Mtb. These findings underscore the importance of using physiologically relevant media in vitro for accurately studying human macrophage function. IMPORTANCE This work compellingly demonstrates that the choice of culture medium significantly influences M. tuberculosis replication outcomes, thus emphasizing the importance of employing physiologically relevant media for accurate in vitro host-pathogen interaction studies. We anticipate that our work will set a precedent for future research with clinical relevance, particularly in evaluating antibiotic efficacy and resistance in cellulo.
Collapse
Affiliation(s)
- Claudio Bussi
- The Francis Crick Institute, London, United Kingdom
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Rachel Lai
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
15
|
Zhang X, Ding G, Yang X, Lu H, Xu Y, Hu Y, Liu S, Zhang H, Huang K, Deng G, Ye T, Yu Q, Cai Y, Xie S, Wang W, Chen X. Myriocin enhances the clearance of M. tuberculosis by macrophages through the activation of PLIN2. mSphere 2024; 9:e0025724. [PMID: 38920406 PMCID: PMC11288015 DOI: 10.1128/msphere.00257-24] [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: 03/27/2024] [Accepted: 05/19/2024] [Indexed: 06/27/2024] Open
Abstract
Myriocin is an inhibitor of de novo synthesis of sphingolipids and ceramides. In this research, we showed myriocin could significantly reduce Mtb burden and histopathological inflammation in mice. However, the underlying mechanism remains unclear. RNA-seq analysis revealed a significant increase in gene expression of PLIN2/CD36/CERT1 after myriocin treatment. The reduced bactericidal burden was only reversed after silencing the lipid droplets (LDs) surface protein PLIN2. This suggests that myriocin enhances the ability of macrophages to clear Mtb depending on the PLIN2 gene, which is part of the PPARγ pathway. Indeed, we observed a significant increase in the number of LDs following myriocin treatment.IMPORTANCEMycobacterium tuberculosis has the ability to reprogram host cell lipid metabolism and alter the antimicrobial functions of infected macrophages. The sphingolipids, such as ceramides, are the primary host lipids utilized by the bacteria, making the sphingomyelinase/ceramide system critical in Mtb infections. Surprisingly, the antimicrobial effect of myriocin was found to be independent of its role in reducing ceramides, but instead, it depends on the lipid droplets surface protein PLIN2. Our findings provide a novel mechanism for how myriocin enhances Mtb clearance in macrophages.
Collapse
Affiliation(s)
- Ximeng Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Guanggui Ding
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xirui Yang
- Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Hailin Lu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yuzhong Xu
- Department of Clinical Laboratory, Shenzhen Baoan Hospital, Shenzhen, China
| | - Yunlong Hu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Song Liu
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Huihua Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Kaisong Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Guofang Deng
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Taosheng Ye
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Qing Yu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Shuixiang Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Wenfei Wang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
16
|
Ting KKY. Revisiting the role of hypoxia-inducible factors and nuclear factor erythroid 2-related factor 2 in regulating macrophage inflammation and metabolism. Front Cell Infect Microbiol 2024; 14:1403915. [PMID: 39119289 PMCID: PMC11306205 DOI: 10.3389/fcimb.2024.1403915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
The recent birth of the immunometabolism field has comprehensively demonstrated how the rewiring of intracellular metabolism is critical for supporting the effector functions of many immune cell types, such as myeloid cells. Among all, the transcriptional regulation mediated by Hypoxia-Inducible Factors (HIFs) and Nuclear factor erythroid 2-related factor 2 (NRF2) have been consistently shown to play critical roles in regulating the glycolytic metabolism, redox homeostasis and inflammatory responses of macrophages (Mφs). Although both of these transcription factors were first discovered back in the 1990s, new advances in understanding their function and regulations have been continuously made in the context of immunometabolism. Therefore, this review attempts to summarize the traditionally and newly identified functions of these transcription factors, including their roles in orchestrating the key events that take place during glycolytic reprogramming in activated myeloid cells, as well as their roles in mediating Mφ inflammatory responses in various bacterial infection models.
Collapse
Affiliation(s)
- Kenneth K. Y. Ting
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
17
|
Wuo MG, Dulberger CL, Warner TC, Brown RA, Sturm A, Ultee E, Bloom-Ackermann Z, Choi C, Zhu J, Garner EC, Briegel A, Hung DT, Rubin EJ, Kiessling LL. Fluorogenic Probes of the Mycobacterial Membrane as Reporters of Antibiotic Action. J Am Chem Soc 2024; 146:17669-17678. [PMID: 38905328 PMCID: PMC11646346 DOI: 10.1021/jacs.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
The genus Mycobacterium includes species such as Mycobacterium tuberculosis, which can cause deadly human diseases. These bacteria have a protective cell envelope that can be remodeled to facilitate their survival in challenging conditions. Understanding how such conditions affect membrane remodeling can facilitate antibiotic discovery and treatment. To this end, we describe an optimized fluorogenic probe, N-QTF, that reports on mycolyltransferase activity, which is vital for cell division and remodeling. N-QTF is a glycolipid probe that can reveal dynamic changes in the mycobacterial cell envelope in both fast- and slow-growing mycobacterial species. Using this probe to monitor the consequences of antibiotic treatment uncovered distinct cellular phenotypes. Even antibiotics that do not directly inhibit cell envelope biosynthesis cause conspicuous phenotypes. For instance, mycobacteria exposed to the RNA polymerase inhibitor rifampicin release fluorescent extracellular vesicles (EVs). While all mycobacteria release EVs, fluorescent EVs were detected only in the presence of RIF, indicating that exposure to the drug alters EV content. Macrophages exposed to the EVs derived from RIF-treated cells released lower levels of cytokines, suggesting the EVs moderate immune responses. These data suggest that antibiotics can alter EV content to impact immunity. Our ability to see such changes in EV constituents directly results from exploiting these chemical probes.
Collapse
Affiliation(s)
- Michael G. Wuo
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue Cambridge, MA 02139, United States
| | - Charles L. Dulberger
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health. 677 Huntington Ave, Boston, MA 02115, United States
- Department of Molecular and Cellular Biology, Harvard University, 52 Oxford St, Cambridge, MA 02138, United States
| | - Theodore C. Warner
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue Cambridge, MA 02139, United States
| | - Robert A. Brown
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue Madison, WI 53706, United States
| | - Alexander Sturm
- Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA 02142, United States
| | - Eveline Ultee
- Institute of Biology, Leiden University, Rapenburg 70, 2311 EZ Leiden, The Netherlands
| | | | - Catherine Choi
- Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA 02142, United States
| | - Junhao Zhu
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health. 677 Huntington Ave, Boston, MA 02115, United States
| | - Ethan C. Garner
- Department of Molecular and Cellular Biology, Harvard University, 52 Oxford St, Cambridge, MA 02138, United States
| | - Ariane Briegel
- Institute of Biology, Leiden University, Rapenburg 70, 2311 EZ Leiden, The Netherlands
| | - Deborah T. Hung
- Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA 02142, United States
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
- Department of Genetics, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, United States
| | - Eric J. Rubin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health. 677 Huntington Ave, Boston, MA 02115, United States
| | - Laura L. Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue Cambridge, MA 02139, United States
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue Madison, WI 53706, United States
| |
Collapse
|
18
|
Hammond FR, Lewis A, Pollara G, Tomlinson GS, Noursadeghi M, Kiss-Toth E, Elks PM. Tribbles1 is host protective during in vivo mycobacterial infection. eLife 2024; 13:e95980. [PMID: 38896446 PMCID: PMC11186633 DOI: 10.7554/elife.95980] [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: 01/10/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Tuberculosis is a major global health problem and is one of the top 10 causes of death worldwide. There is a pressing need for new treatments that circumvent emerging antibiotic resistance. Mycobacterium tuberculosis parasitises macrophages, reprogramming them to establish a niche in which to proliferate, therefore macrophage manipulation is a potential host-directed therapy if druggable molecular targets could be identified. The pseudokinase Tribbles1 (Trib1) regulates multiple innate immune processes and inflammatory profiles making it a potential drug target in infections. Trib1 controls macrophage function, cytokine production, and macrophage polarisation. Despite wide-ranging effects on leukocyte biology, data exploring the roles of Tribbles in infection in vivo are limited. Here, we identify that human Tribbles1 is expressed in monocytes and is upregulated at the transcript level after stimulation with mycobacterial antigen. To investigate the mechanistic roles of Tribbles in the host response to mycobacteria in vivo, we used a zebrafish Mycobacterium marinum (Mm) infection tuberculosis model. Zebrafish Tribbles family members were characterised and shown to have substantial mRNA and protein sequence homology to their human orthologues. trib1 overexpression was host-protective against Mm infection, reducing burden by approximately 50%. Conversely, trib1 knockdown/knockout exhibited increased infection. Mechanistically, trib1 overexpression significantly increased the levels of proinflammatory factors il-1β and nitric oxide. The host-protective effect of trib1 was found to be dependent on the E3 ubiquitin kinase Cop1. These findings highlight the importance of Trib1 and Cop1 as immune regulators during infection in vivo and suggest that enhancing macrophage TRIB1 levels may provide a tractable therapeutic intervention to improve bacterial infection outcomes in tuberculosis.
Collapse
Affiliation(s)
- Ffion R Hammond
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Amy Lewis
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Gabriele Pollara
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Gillian S Tomlinson
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Mahdad Noursadeghi
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Endre Kiss-Toth
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Philip M Elks
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
19
|
Vu A, Glassman I, Campbell G, Yeganyan S, Nguyen J, Shin A, Venketaraman V. Host Cell Death and Modulation of Immune Response against Mycobacterium tuberculosis Infection. Int J Mol Sci 2024; 25:6255. [PMID: 38892443 PMCID: PMC11172987 DOI: 10.3390/ijms25116255] [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: 05/01/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a prevalent infectious disease affecting populations worldwide. A classic trait of TB pathology is the formation of granulomas, which wall off the pathogen, via the innate and adaptive immune systems. Some key players involved include tumor necrosis factor-alpha (TNF-α), foamy macrophages, type I interferons (IFNs), and reactive oxygen species, which may also show overlap with cell death pathways. Additionally, host cell death is a primary method for combating and controlling Mtb within the body, a process which is influenced by both host and bacterial factors. These cell death modalities have distinct molecular mechanisms and pathways. Programmed cell death (PCD), encompassing apoptosis and autophagy, typically confers a protective response against Mtb by containing the bacteria within dead macrophages, facilitating their phagocytosis by uninfected or neighboring cells, whereas necrotic cell death benefits the pathogen, leading to the release of bacteria extracellularly. Apoptosis is triggered via intrinsic and extrinsic caspase-dependent pathways as well as caspase-independent pathways. Necrosis is induced via various pathways, including necroptosis, pyroptosis, and ferroptosis. Given the pivotal role of host cell death pathways in host defense against Mtb, therapeutic agents targeting cell death signaling have been investigated for TB treatment. This review provides an overview of the diverse mechanisms underlying Mtb-induced host cell death, examining their implications for host immunity. Furthermore, it discusses the potential of targeting host cell death pathways as therapeutic and preventive strategies against Mtb infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (G.C.); (A.S.)
| |
Collapse
|
20
|
Li C, Wang J, Xu JF, Pi J, Zheng B. Roles of HIF-1α signaling in Mycobacterium tuberculosis infection: New targets for anti-TB therapeutics? Biochem Biophys Res Commun 2024; 711:149920. [PMID: 38615574 DOI: 10.1016/j.bbrc.2024.149920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Tuberculosis (TB), a deadly infectious disease induced by Mycobacterium tuberculosis (Mtb), continues to be a global public health issue that kill millions of patents every year. Despite significant efforts have been paid to identify effective TB treatments, the emergence of drug-resistant strains of the disease and the presence of comorbidities in TB patients urges us to explore the detailed mechanisms involved in TB immunity and develop more effective innovative anti-TB strategies. HIF-1α, a protein involved in regulating cellular immune responses during TB infection, has been highlighted as a promising target for the development of novel strategies for TB treatment due to its critical roles in anti-TB host immunity. This review provides a summary of current research progress on the roles of HIF-1α in TB infection, highlighting its importance in regulating the host immune response upon Mtb infection and summarizing the influences and mechanisms of HIF-1α on anti-TB immunological responses of host cells. This review also discusses the various challenges associated with developing HIF-1α as a target for anti-TB therapies, including ensuring specificity and avoiding off-target effects on normal cell function, determining the regulation and expression of HIF-1α in TB patients, and developing drugs that can inhibit HIF-1α. More deep understanding of the molecular mechanisms involved in HIF-1α signaling, its impact on TB host status, and systematic animal testing and clinical trials may benefit the optimization of HIF-1α as a novel therapeutic target for TB.
Collapse
Affiliation(s)
- Chaowei Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jiajun Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Biying Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| |
Collapse
|
21
|
Piccaro G, Aquino G, Gigantino V, Tirelli V, Sanchez M, Iorio E, Matarese G, Cassone A, Palma C. Mycobacterium tuberculosis antigen 85B modifies BCG-induced antituberculosis immunity and favors pathogen survival. J Leukoc Biol 2024; 115:1053-1069. [PMID: 38242866 DOI: 10.1093/jleuko/qiae014] [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: 04/20/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Tuberculosis is one of the deadliest infectious diseases worldwide. Mycobacterium tuberculosis has developed strategies not only to evade host immunity but also to manipulate it for its survival. We investigated whether Mycobacterium tuberculosis exploited the immunogenicity of Ag85B, one of its major secretory proteins, to redirect host antituberculosis immunity to its advantage. We found that administration of Ag85B protein to mice vaccinated with Bacillus Calmette-Guérin impaired the protection elicited by vaccination, causing a more severe infection when mice were challenged with Mycobacterium tuberculosis. Ag85B administration reduced Bacillus Calmette-Guérin-induced CD4 T-cell activation and IFN-γ, CCL-4, and IL-22 production in response to Mycobacterium tuberculosis-infected cells. On the other hand, it promoted robust Ag85B-responsive IFN-γ-producing CD4 T cells, expansion of a subset of IFN-γ/IL-10-producing CD4+FOXP3+Treg cells, differential activation of IL-17/IL-22 responses, and activation of regulatory and exhaustion pathways, including programmed death ligand 1 expression on macrophages. All this resulted in impaired intracellular Mycobacterium tuberculosis growth control by systemic immunity, both before and after the Mycobacterium tuberculosis challenge. Interestingly, Mycobacterium tuberculosis infection itself generated Ag85B-reactive inflammatory immune cells incapable of clearing Mycobacterium tuberculosis in both unvaccinated and Bacillus Calmette-Guérin-vaccinated mice. Our data suggest that Mycobacterium tuberculosis can exploit the strong immunogenicity of Ag85B to promote its own survival and spread. Since Ag85B is normally secreted by replicating bacteria and is commonly found in the lungs of the Mycobacterium tuberculosis-infected host, our findings may advance the understanding on the mechanisms of Mycobacterium tuberculosis pathogenesis and immune evasion.
Collapse
Affiliation(s)
- Giovanni Piccaro
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Gabriella Aquino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Valentina Tirelli
- Core Facilities-Flow Cytometry Area, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Massimo Sanchez
- Core Facilities-Flow Cytometry Area, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Egidio Iorio
- Core Facilities-High Resolution NMR Unit, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie mediche, Università di Napoli "Federico II," Via Sergio Pansini 5, 80131 Naples, Italy
| | - Antonio Cassone
- Polo d'innovazione della Genomica, Genetica e Biologia, Via Fiorentina 1, 53100 Siena, Italy
| | - Carla Palma
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
22
|
Wiggins DA, Maxwell JN, Nelson DE. Exploring the role of CITED transcriptional regulators in the control of macrophage polarization. Front Immunol 2024; 15:1365718. [PMID: 38646545 PMCID: PMC11032013 DOI: 10.3389/fimmu.2024.1365718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Macrophages are tissue resident innate phagocytic cells that take on contrasting phenotypes, or polarization states, in response to the changing combination of microbial and cytokine signals at sites of infection. During the opening stages of an infection, macrophages adopt the proinflammatory, highly antimicrobial M1 state, later shifting to an anti-inflammatory, pro-tissue repair M2 state as the infection resolves. The changes in gene expression underlying these transitions are primarily governed by nuclear factor kappaB (NF-κB), Janus kinase (JAK)/signal transducer and activation of transcription (STAT), and hypoxia-inducible factor 1 (HIF1) transcription factors, the activity of which must be carefully controlled to ensure an effective yet spatially and temporally restricted inflammatory response. While much of this control is provided by pathway-specific feedback loops, recent work has shown that the transcriptional co-regulators of the CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxy-terminal domain (CITED) family serve as common controllers for these pathways. In this review, we describe how CITED proteins regulate polarization-associated gene expression changes by controlling the ability of transcription factors to form chromatin complexes with the histone acetyltransferase, CBP/p300. We will also cover how differences in the interactions between CITED1 and 2 with CBP/p300 drive their contrasting effects on pro-inflammatory gene expression.
Collapse
Affiliation(s)
| | | | - David E. Nelson
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, United States
| |
Collapse
|
23
|
Tan YJ, Jin Y, Zhou J, Yang YF. Lipid droplets in pathogen infection and host immunity. Acta Pharmacol Sin 2024; 45:449-464. [PMID: 37993536 PMCID: PMC10834987 DOI: 10.1038/s41401-023-01189-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
As the hub of cellular lipid metabolism, lipid droplets (LDs) have been linked to a variety of biological processes. During pathogen infection, the biogenesis, composition, and functions of LDs are tightly regulated. The accumulation of LDs has been described as a hallmark of pathogen infection and is thought to be driven by pathogens for their own benefit. Recent studies have revealed that LDs and their subsequent lipid mediators contribute to effective immunological responses to pathogen infection by promoting host stress tolerance and reducing toxicity. In this comprehensive review, we delve into the intricate roles of LDs in governing the replication and assembly of a wide spectrum of pathogens within host cells. We also discuss the regulatory function of LDs in host immunity and highlight the potential for targeting LDs for the diagnosis and treatment of infectious diseases.
Collapse
Affiliation(s)
- Yan-Jie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Yi Jin
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yun-Fan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
24
|
Sood C, Verma JK, Basak R, Kapoor A, Gupta S, Mukhopadhyay A. Leishmania highjack host lipid body for its proliferation in macrophages by overexpressing host Rab18 and TRAPPC9 by downregulating miR-1914-3p expression. PLoS Pathog 2024; 20:e1012024. [PMID: 38412149 PMCID: PMC10898768 DOI: 10.1371/journal.ppat.1012024] [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: 04/29/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024] Open
Abstract
Lipids stored in lipid-bodies (LBs) in host cells are potential sources of fatty acids for pathogens. However, the mechanism of recruitment of LBs from the host cells by pathogens to acquire fatty acids is not known. Here, we have found that Leishmania specifically upregulates the expression of host Rab18 and its GEF, TRAPPC9 by downregulating the expression of miR-1914-3p by reducing the level of Dicer in macrophages via their metalloprotease gp63. Our results also show that miR-1914-3p negatively regulates the expression of Rab18 and its GEF in cells. Subsequently, Leishmania containing parasitophorous vacuoles (Ld-PVs) recruit and retain host Rab18 and TRAPPC9. Leishmania infection also induces LB biogenesis in host cells and recruits LBs on Ld-PVs and acquires FLC12-labeled fatty acids from LBs. Moreover, overexpression of miR-1914-3p in macrophages significantly inhibits the recruitment of LBs and thereby suppresses the multiplication of parasites in macrophages as parasites are unable to acquire fatty acids. These results demonstrate a novel mechanism how Leishmania acquire fatty acids from LBs for their growth in macrophages.
Collapse
Affiliation(s)
- Chandni Sood
- National Institute of Immunology, New Delhi, India
| | - Jitender Kumar Verma
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
- National Institute of Immunology, New Delhi, India
| | - Rituparna Basak
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Anjali Kapoor
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Swarnima Gupta
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Amitabha Mukhopadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
- National Institute of Immunology, New Delhi, India
| |
Collapse
|
25
|
Chugh S, Tiwari P, Suri C, Gupta SK, Singh P, Bouzeyen R, Kidwai S, Srivastava M, Rameshwaram NR, Kumar Y, Asthana S, Singh R. Polyphosphate kinase-1 regulates bacterial and host metabolic pathways involved in pathogenesis of Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 2024; 121:e2309664121. [PMID: 38170746 PMCID: PMC10786269 DOI: 10.1073/pnas.2309664121] [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: 06/18/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
Inorganic polyphosphate (polyP) is primarily synthesized by Polyphosphate Kinase-1 (PPK-1) and regulates numerous cellular processes, including energy metabolism, stress adaptation, drug tolerance, and microbial pathogenesis. Here, we report that polyP interacts with acyl CoA carboxylases, enzymes involved in lipid biosynthesis in Mycobacterium tuberculosis. We show that deletion of ppk-1 in M. tuberculosis results in transcriptional and metabolic reprogramming. In comparison to the parental strain, the Δppk-1 mutant strain had reduced levels of virulence-associated lipids such as PDIMs and TDM. We also observed that polyP deficiency in M. tuberculosis is associated with enhanced phagosome-lysosome fusion in infected macrophages and attenuated growth in mice. Host RNA-seq analysis revealed decreased levels of transcripts encoding for proteins involved in either type I interferon signaling or formation of foamy macrophages in the lungs of Δppk-1 mutant-infected mice relative to parental strain-infected animals. Using target-based screening and molecular docking, we have identified raloxifene hydrochloride as a broad-spectrum PPK-1 inhibitor. We show that raloxifene hydrochloride significantly enhanced the activity of isoniazid, bedaquiline, and pretomanid against M. tuberculosis in macrophages. Additionally, raloxifene inhibited the growth of M. tuberculosis in mice. This is an in-depth study that provides mechanistic insights into the regulation of mycobacterial pathogenesis by polyP deficiency.
Collapse
Affiliation(s)
- Saurabh Chugh
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Prabhakar Tiwari
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Charu Suri
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Sonu Kumar Gupta
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Padam Singh
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Rania Bouzeyen
- Institut Pasteur de Tunis, Laboratory of Transmission, Control and Immunobiology of Infections, LRII IPT02, Tunis1002, Tunisia
| | - Saqib Kidwai
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Mitul Srivastava
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Nagender Rao Rameshwaram
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Yashwant Kumar
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Shailendra Asthana
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Ramandeep Singh
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| |
Collapse
|
26
|
Serio B, Giudice V, Selleri C. All Roads Lead to Interferon-γ: From Known to Untraveled Pathways in Acquired Aplastic Anemia. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2170. [PMID: 38138273 PMCID: PMC10744863 DOI: 10.3390/medicina59122170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
Bone marrow failure (BMF) syndromes are a heterogeneous group of benign hematological conditions with common clinical features including reduced bone marrow cellularity and peripheral blood cytopenias. Acquired aplastic anemia (AA) is caused by T helper(Th)1-mediated immune responses and cytotoxic CD8+ T cell-mediated autologous immune attacks against hematopoietic stem and progenitor cells (HSPCs). Interferon-γ (IFNγ), tumor necrosis factor-α, and Fas-ligand are historically linked to AA pathogenesis because they drive Th1 and cytotoxic T cell-mediated responses and can directly induce HSPC apoptosis and differentiation block. The use of omics technologies has amplified the amount of data at the single-cell level, and knowledge on AA, and new scenarios, have been opened on "old" point of view. In this review, we summarize the current state-of-art of the pathogenic role of IFNγ in AA from initial findings to novel evidence, such as the involvement of the HIF-1α pathway, and how this knowledge can be translated in clinical practice.
Collapse
Affiliation(s)
- Bianca Serio
- Department of Medicine, Surgery, and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (B.S.); (C.S.)
| | - Valentina Giudice
- Department of Medicine, Surgery, and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (B.S.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery, and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (B.S.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
27
|
Challagundla N, Phadnis D, Gupta A, Agrawal-Rajput R. Host Lipid Manipulation by Intracellular Bacteria: Moonlighting for Immune Evasion. J Membr Biol 2023; 256:393-411. [PMID: 37938349 DOI: 10.1007/s00232-023-00296-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023]
Abstract
Lipids are complex organic molecules that fulfill energy demands and sometimes act as signaling molecules. They are mostly found in membranes, thus playing an important role in membrane trafficking and protecting the cell from external dangers. Based on the composition of the lipids, their fluidity and charge, their interaction with embedded proteins vary greatly. Bacteria can hijack host lipids to satisfy their energy needs or to conceal themselves from host cells. Intracellular bacteria continuously exploit host, from their entry into host cells utilizing host lipid machinery to exiting through the cells. This acquisition of lipids from host cells helps in their disguise mechanism. The current review explores various mechanisms employed by the intracellular bacteria to manipulate and acquire host lipids. It discusses their role in manipulating host membranes and the subsequence impact on the host cells. Modulating these lipids in macrophages not only serve the purpose of the pathogen but also modulates the macrophage energy metabolism and functional state. Additionally, we have explored the intricate pathogenic relationship and the potential prospects of using this knowledge in lipid-based therapeutics to disrupt pathogen dominance.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat, 382426, India
| | - Deepti Phadnis
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat, 382426, India
| | - Aakriti Gupta
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat, 382426, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat, 382426, India.
| |
Collapse
|
28
|
Ni D, Zhou H, Wang P, Xu F, Li C. Visualizing Macrophage Phenotypes and Polarization in Diseases: From Biomarkers to Molecular Probes. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:613-638. [PMID: 38223685 PMCID: PMC10781933 DOI: 10.1007/s43657-023-00129-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 01/16/2024]
Abstract
Macrophage is a kind of immune cell and performs multiple functions including pathogen phagocytosis, antigen presentation and tissue remodeling. To fulfill their functionally distinct roles, macrophages undergo polarization towards a spectrum of phenotypes, particularly the classically activated (M1) and alternatively activated (M2) subtypes. However, the binary M1/M2 phenotype fails to capture the complexity of macrophages subpopulations in vivo. Hence, it is crucial to employ spatiotemporal imaging techniques to visualize macrophage phenotypes and polarization, enabling the monitoring of disease progression and assessment of therapeutic responses to drug candidates. This review begins by discussing the origin, function and diversity of macrophage under physiological and pathological conditions. Subsequently, we summarize the identified macrophage phenotypes and their specific biomarkers. In addition, we present the imaging probes locating the lesions by visualizing macrophages with specific phenotype in vivo. Finally, we discuss the challenges and prospects associated with monitoring immune microenvironment and disease progression through imaging of macrophage phenotypes.
Collapse
Affiliation(s)
- Dan Ni
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
| | - Heqing Zhou
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Pengwei Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
| | - Fulin Xu
- Minhang Hospital, Fudan University, Shanghai, 201199 China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 201203 China
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, 201203 China
| |
Collapse
|
29
|
Madduri BTSA, Bell SL. Bug in the code: TB blocks DNA repair. Cell Host Microbe 2023; 31:1769-1771. [PMID: 37944488 DOI: 10.1016/j.chom.2023.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
Protecting the cell's genome is crucial for survival, but infection causes damage that compromises genetic integrity. In this issue of Cell Host & Microbe, Lui et al. dissect how Mycobacterium tuberculosis exploits DNA damage using a secreted protein that inhibits DNA repair to create an environment conducive to bacterial replication.
Collapse
Affiliation(s)
- Bala T S A Madduri
- Center for Emerging & Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, USA; Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Samantha L Bell
- Center for Emerging & Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, USA; Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
30
|
Prado LG, Camara NOS, Barbosa AS. Cell lipid biology in infections: an overview. Front Cell Infect Microbiol 2023; 13:1148383. [PMID: 37868347 PMCID: PMC10587689 DOI: 10.3389/fcimb.2023.1148383] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Lipids are a big family of molecules with a vast number of functions in the cell membranes, within the cytoplasm, and extracellularly. Lipid droplets (LDs) are the most common storage organelles and are present in almost every tissue type in the body. They also have structural functions serving as building blocks of cellular membranes and may be precursors of other molecules such as hormones, and lipoproteins, and as messengers in signal transduction. Fatty acids (FAs), such as sterol esters and triacylglycerols, are stored in LDs and are used in β-oxidation as fuel for tricarboxylic acid cycle (TCA) and adenosine triphosphate (ATP) generation. FA uptake and entrance in the cytoplasm are mediated by membrane receptors. After a cytoplasmic round of α- and β-oxidation, FAs are guided into the mitochondrial matrix by the L-carnitine shuttle system, where they are fully metabolized, and enter the TCA cycle. Pathogen infections may lead to impaired lipid metabolism, usage of membrane phospholipids, and LD accumulation in the cytoplasm of infected cells. Otherwise, bacterial pathogens may use lipid metabolism as a carbon source, thus altering the reactions and leading to cellular and organelles malfunctioning. This review aims to describe cellular lipid metabolism and alterations that occur upon infections.
Collapse
Affiliation(s)
- Luan Gavião Prado
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Camara
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
31
|
Kim H, Shin SJ. Revolutionizing control strategies against Mycobacterium tuberculosis infection through selected targeting of lipid metabolism. Cell Mol Life Sci 2023; 80:291. [PMID: 37704889 PMCID: PMC11072447 DOI: 10.1007/s00018-023-04914-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/12/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023]
Abstract
Lipid species play a critical role in the growth and virulence expression of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). During Mtb infection, foamy macrophages accumulate lipids in granulomas, providing metabolic adaptation and survival strategies for Mtb against multiple stresses. Host-derived lipid species, including triacylglycerol and cholesterol, can also contribute to the development of drug-tolerant Mtb, leading to reduced efficacy of antibiotics targeting the bacterial cell wall or transcription. Transcriptional and metabolic analyses indicate that lipid metabolism-associated factors of Mtb are highly regulated by antibiotics and ultimately affect treatment outcomes. Despite the well-known association between major antibiotics and lipid metabolites in TB treatment, a comprehensive understanding of how altered lipid metabolites in both host and Mtb influence treatment outcomes in a drug-specific manner is necessary to overcome drug tolerance. The current review explores the controversies and correlations between lipids and drug efficacy in various Mtb infection models and proposes novel approaches to enhance the efficacy of anti-TB drugs. Moreover, the review provides insights into the efficacious control of Mtb infection by elucidating the impact of lipids on drug efficacy. This review aims to improve the effectiveness of current anti-TB drugs and facilitate the development of innovative therapeutic strategies against Mtb infection by making reverse use of Mtb-favoring lipid species.
Collapse
Affiliation(s)
- Hagyu Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
32
|
Roy A, Kumari Agnivesh P, Sau S, Kumar S, Pal Kalia N. Tweaking host immune responses for novel therapeutic approaches against Mycobacterium tuberculosis. Drug Discov Today 2023; 28:103693. [PMID: 37390961 DOI: 10.1016/j.drudis.2023.103693] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023]
Abstract
In TB, combat between the human host and Mycobacterium tuberculosis involves intricate interactions with immune cells. M. tuberculosis has evolved a complex evasion system to circumvent immune cells, leading to persistence and limiting its clearance by the host. Host-directed therapies are emerging approaches to modulate host responses, including inflammatory responses, cytokine responses, and autophagy, by using small molecules to curb mycobacterial infections. Targeting host immune pathways reduces the chances of antibiotic resistance to M. tuberculosis because, unlike antibiotics, this approach acts directly on the cells of the host. In this review, we discuss the role of immune cells during M. tuberculosis proliferation, provide a updated understanding of immunopathogenesis, and explore the range of host-modulating options for the clearance of this pathogen.
Collapse
Affiliation(s)
- Arnab Roy
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500 037, India
| | - Puja Kumari Agnivesh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500 037, India
| | - Shashikanta Sau
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500 037, India
| | - Sunil Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500 037, India
| | - Nitin Pal Kalia
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500 037, India.
| |
Collapse
|
33
|
Fu S, Wang Z, Han X, Xu Y, Miao J. The therapeutic potential for targeting CSE/H 2S signaling in macrophages against Escherichia coli infection. Vet Res 2023; 54:71. [PMID: 37644526 PMCID: PMC10466716 DOI: 10.1186/s13567-023-01203-8] [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: 05/11/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
Macrophages play a pivotal role in the inflammatory response to the zoonotic pathogen E. coli, responsible for causing enteric infections. While considerable research has been conducted to comprehend the pathogenesis of this disease, scant attention devoted to host-derived H2S. Herein, we reported that E. coli infection enhanced the expression of CSE in macrophages, accompanied by a significantly increased inflammatory response. This process may be mediated by the involvement of excessive autophagy. Inhibition of AMPK or autophagy with pharmacological inhibitors could alleviate the inflammation. Additionally, cell model showed that the mRNA expression of classic inflammatory factors (Il-1β, Il-6), macrophage polarization markers (iNOS, Arg1) and ROS production was significantly down-regulated after employing CSE specific inhibitor PAG. And PAG is capable of inhibiting excessive autophagy through the LKB1-AMPK-ULK1 axis. Interestingly, exogenous H2S could suppress inflammation response. Our study emphasizes the importance of CSE in regulating the macrophage-mediated response to E. coli. Increased CSE in macrophages leads to excessive inflammation, which should be considered a new target for drug development to treat intestinal infection.
Collapse
Affiliation(s)
- Shaodong Fu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhenglei Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
34
|
Fines DM, Schichnes D, Knight M, Anaya-Sanchez A, Thuong N, Cox J, Stanley SA. Mycobacterial formation of intracellular lipid inclusions is a dynamic process associated with rapid replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552809. [PMID: 37609245 PMCID: PMC10441389 DOI: 10.1101/2023.08.10.552809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Intracellular lipid inclusions (ILI) are triacylglyceride rich organelles produced by mycobacteria thought to serve as energy reservoirs. It is believed that ILI are formed as a result of a dosR mediated transition from replicative growth to non-replicating persistence (NRP). ILI rich Mycobacterium tuberculosis (Mtb) bacilli have been reported during infection and in sputum, establishing their importance in Mtb pathogenesis. Studies conducted in mycobacteria such as Mycobacterium smegmatis, Mycobacterium abscessus, or lab Mtb strains have demonstrated ILI formation in the presence of hypoxic, nitric oxide, nutrient limitation, or low nitrogen stress, conditions believed to emulate the host environment within which Mtb resides. Here, we show that M. marinum and clinical Mtb isolates make ILI during active replication in axenic culture independent of environmental stressors. By tracking ILI formation dynamics we demonstrate that ILI are quickly formed in the presence of fresh media or exogenous fatty acids but are rapidly depleted while bacteria are still actively replicating. We also show that the cell envelope is an alternate site for neutral lipid accumulation observed during stationary phase. In addition, we screen a panel of 60 clinical isolates and observe variation in ILI production during early log phase growth between and among Mtb lineages. Finally, we show that dosR expression level does not strictly correlate with ILI accumulation in fresh clinical isolates. Taken together, our data provide evidence of an active ILI formation pathway in replicating mycobacteria cultured in the absence of stressors, suggesting a decoupling of ILI formation from NRP.
Collapse
|
35
|
Safi R, Sánchez-Álvarez M, Bosch M, Demangel C, Parton RG, Pol A. Defensive-lipid droplets: Cellular organelles designed for antimicrobial immunity. Immunol Rev 2023; 317:113-136. [PMID: 36960679 DOI: 10.1111/imr.13199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
Microbes have developed many strategies to subvert host organisms, which, in turn, evolved several innate immune responses. As major lipid storage organelles of eukaryotes, lipid droplets (LDs) are an attractive source of nutrients for invaders. Intracellular viruses, bacteria, and protozoan parasites induce and physically interact with LDs, and the current view is that they "hijack" LDs to draw on substrates for host colonization. This dogma has been challenged by the recent demonstration that LDs are endowed with a protein-mediated antibiotic activity, which is upregulated in response to danger signals and sepsis. Dependence on host nutrients could be a generic "Achilles' heel" of intracellular pathogens and LDs a suitable chokepoint harnessed by innate immunity to organize a front-line defense. Here, we will provide a brief overview of the state of the conflict and discuss potential mechanisms driving the formation of the 'defensive-LDs' functioning as hubs of innate immunity.
Collapse
Affiliation(s)
- Rémi Safi
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Miguel Sánchez-Álvarez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (IIB), Madrid, Spain
| | - Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Caroline Demangel
- Immunobiology and Therapy Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Robert G Parton
- Institute for Molecular Bioscience (IMB), Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis (CMM), University of Queensland, Brisbane, Queensland, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
36
|
Hüsler D, Stauffer P, Hilbi H. Tapping lipid droplets: A rich fat diet of intracellular bacterial pathogens. Mol Microbiol 2023; 120:194-209. [PMID: 37429596 DOI: 10.1111/mmi.15120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023]
Abstract
Lipid droplets (LDs) are dynamic and versatile organelles present in most eukaryotic cells. LDs consist of a hydrophobic core of neutral lipids, a phospholipid monolayer coat, and a variety of associated proteins. LDs are formed at the endoplasmic reticulum and have diverse roles in lipid storage, energy metabolism, membrane trafficking, and cellular signaling. In addition to their physiological cellular functions, LDs have been implicated in the pathogenesis of several diseases, including metabolic disorders, cancer, and infections. A number of intracellular bacterial pathogens modulate and/or interact with LDs during host cell infection. Members of the genera Mycobacterium, Legionella, Coxiella, Chlamydia, and Salmonella exploit LDs as a source of intracellular nutrients and membrane components to establish their distinct intracellular replicative niches. In this review, we focus on the biogenesis, interactions, and functions of LDs, as well as on their role in lipid metabolism of intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Dario Hüsler
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Pia Stauffer
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Gonçalves LED, Andrade-Silva M, Basso PJ, Câmara NOS. Vitamin D and chronic kidney disease: Insights on lipid metabolism of tubular epithelial cell and macrophages in tubulointerstitial fibrosis. Front Physiol 2023; 14:1145233. [PMID: 37064892 PMCID: PMC10090472 DOI: 10.3389/fphys.2023.1145233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Chronic kidney disease (CKD) has been recognized as a significant global health problem due to being an important contributor to morbidity and mortality. Inflammation is the critical event that leads to CKD development orchestrated by a complex interaction between renal parenchyma and immune cells. Particularly, the crosstalk between tubular epithelial cells (TECs) and macrophages is an example of the critical cell communication in the kidney that drives kidney fibrosis, a pathological feature in CKD. Metabolism dysregulation of TECs and macrophages can be a bridge that connects inflammation and fibrogenesis. Currently, some evidence has reported how cellular lipid disturbances can affect kidney disease and cause tubulointerstitial fibrosis highlighting the importance of investigating potential molecules that can restore metabolic parameters. Vitamin D (VitD) is a hormone naturally produced by mammalian cells in a coordinated manner by the skin, liver, and kidneys. VitD deficiency or insufficiency is prevalent in patients with CKD, and serum levels of VitD are inversely correlated with the degree of kidney inflammation and renal function. Proximal TECs and macrophages produce the active form of VitD, and both express the VitD receptor (VDR) that evidence the importance of this nutrient in regulating their functions. However, whether VitD signaling drives physiological and metabolism improvement of TECs and macrophages during kidney injury is an open issue to be debated. In this review, we brought to light VitD as an important metabolic modulator of lipid metabolism in TECs and macrophages. New scientific approaches targeting VitD e VDR signaling at the cellular metabolic level can provide a better comprehension of its role in renal physiology and CKD progression.
Collapse
Affiliation(s)
- Luís Eduardo D. Gonçalves
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Magaiver Andrade-Silva
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Laboratory of Experimental e Clinical Immunology, Department of Clinical Medicine, Faculty of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Paulo José Basso
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- *Correspondence: Paulo José Basso, ; Niels O. S. Câmara,
| | - Niels O. S. Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Laboratory of Experimental e Clinical Immunology, Department of Clinical Medicine, Faculty of Medicine, Federal University of São Paulo, São Paulo, Brazil
- *Correspondence: Paulo José Basso, ; Niels O. S. Câmara,
| |
Collapse
|
38
|
Rombouts Y, Neyrolles O. The fat is in the lysosome: how Mycobacterium tuberculosis tricks macrophages into storing lipids. J Clin Invest 2023; 133:168366. [PMID: 36919697 PMCID: PMC10014092 DOI: 10.1172/jci168366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), infects primarily macrophages, causing them to differentiate into lipid-laden foamy macrophages that are a primary source of tissue destruction in patients with TB. In this issue of the JCI, Bedard et al. demonstrate that 1-tuberculosinyladenosine, a virulence factor produced by M. tuberculosis, caused lysosomal dysfunction associated with lipid storage in the phagolysosome of macrophages in a manner that mimicked lysosomal storage diseases. This work sheds light on how M. tuberculosis manipulates host lipid metabolism for its survival and opens avenues toward host-directed therapy against TB.
Collapse
|
39
|
Lanni F, Wijnant GJ, Xie M, Osiecki P, Dartois V, Sarathy JP. Adaptation to the intracellular environment of primary human macrophages influences drug susceptibility of Mycobacterium tuberculosis. Tuberculosis (Edinb) 2023; 139:102318. [PMID: 36889104 DOI: 10.1016/j.tube.2023.102318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 01/24/2023]
Abstract
As a facultative intracellular pathogen, M. tuberculosis (Mtb) is highly adapted to evading antibacterial mechanisms in phagocytic cells. Both the macrophage and pathogen experience transcriptional and metabolic changes from the onset of phagocytosis. To account for this interaction in the assessment of intracellular drug susceptibility, we allowed a 3-day preadaptation phase post-macrophage infection prior to drug treatment. We found that intracellular Mtb in human monocyte-derived macrophages (MDM) presents dramatic alterations in susceptibility to isoniazid, sutezolid, rifampicin and rifapentine when compared to axenic culture. Infected MDM gradually accumulate lipid bodies, adopting a characteristic appearance reminiscent of foamy macrophages in granulomas. Furthermore, TB granulomas in vivo develop hypoxic cores with decreasing oxygen tension gradients across their radii. Accordingly, we evaluated the effects of hypoxia on preadapted intracellular Mtb in our MDM model. We observed that hypoxia induced greater lipid body formation and no additional shifts in drug tolerance, suggesting that the adaptation of intracellular Mtb to baseline host cell conditions under normoxia dominates changes to intracellular drug susceptibility. Using unbound plasma concentrations in patients as surrogates for free drug concentrations in lung interstitial fluid, we estimate that intramacrophage Mtb in granulomas are exposed to bacteriostatic concentrations of most study drugs.
Collapse
Affiliation(s)
- Faye Lanni
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Gert-Jan Wijnant
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Min Xie
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Paulina Osiecki
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States; Hackensack School of Medicine, Department of Medical Sciences, 123, Metro Boulevard, Nutley, NJ, 07110, United States
| | - Jansy P Sarathy
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States.
| |
Collapse
|
40
|
Belhaouane I, Pochet A, Chatagnon J, Hoffmann E, Queval CJ, Deboosère N, Boidin-Wichlacz C, Majlessi L, Sencio V, Heumel S, Vandeputte A, Werkmeister E, Fievez L, Bureau F, Rouillé Y, Trottein F, Chamaillard M, Brodin P, Machelart A. Tirap controls Mycobacterium tuberculosis phagosomal acidification. PLoS Pathog 2023; 19:e1011192. [PMID: 36888688 PMCID: PMC9994722 DOI: 10.1371/journal.ppat.1011192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/30/2023] [Indexed: 03/09/2023] Open
Abstract
Progression of tuberculosis is tightly linked to a disordered immune balance, resulting in inability of the host to restrict intracellular bacterial replication and its subsequent dissemination. The immune response is mainly characterized by an orchestrated recruitment of inflammatory cells secreting cytokines. This response results from the activation of innate immunity receptors that trigger downstream intracellular signaling pathways involving adaptor proteins such as the TIR-containing adaptor protein (Tirap). In humans, resistance to tuberculosis is associated with a loss-of-function in Tirap. Here, we explore how genetic deficiency in Tirap impacts resistance to Mycobacterium tuberculosis (Mtb) infection in a mouse model and ex vivo. Interestingly, compared to wild type littermates, Tirap heterozygous mice were more resistant to Mtb infection. Upon investigation at the cellular level, we observed that mycobacteria were not able to replicate in Tirap-deficient macrophages compared to wild type counterparts. We next showed that Mtb infection induced Tirap expression which prevented phagosomal acidification and rupture. We further demonstrate that the Tirap-mediated anti-tuberculosis effect occurs through a Cish-dependent signaling pathway. Our findings provide new molecular evidence about how Mtb manipulates innate immune signaling to enable intracellular replication and survival of the pathogen, thus paving the way for host-directed approaches to treat tuberculosis.
Collapse
Affiliation(s)
- Imène Belhaouane
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Amine Pochet
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Jonathan Chatagnon
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Eik Hoffmann
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Christophe J. Queval
- High Throughput Screening Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Nathalie Deboosère
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Céline Boidin-Wichlacz
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, Paris, France
| | - Valentin Sencio
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Séverine Heumel
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Alexandre Vandeputte
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Elisabeth Werkmeister
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UMS 2014—PLBS, Lille, France
| | - Laurence Fievez
- Laboratory of Cellular and Molecular Immunology, GIGA-Research, Liège, Belgium
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, GIGA-Research, Liège, Belgium
| | - Yves Rouillé
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - François Trottein
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Mathias Chamaillard
- Laboratory of Cell Physiology, INSERM U1003, University of Lille, Lille, France
| | - Priscille Brodin
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
- * E-mail: (PB); (AM)
| | - Arnaud Machelart
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
- * E-mail: (PB); (AM)
| |
Collapse
|
41
|
Arbaizar-Rovirosa M, Pedragosa J, Lozano JJ, Casal C, Pol A, Gallizioli M, Planas AM. Aged lipid-laden microglia display impaired responses to stroke. EMBO Mol Med 2023; 15:e17175. [PMID: 36541061 PMCID: PMC9906381 DOI: 10.15252/emmm.202217175] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Microglial cells of the aged brain manifest signs of dysfunction that could contribute to the worse neurological outcome of stroke in the elderly. Treatment with colony-stimulating factor 1 receptor antagonists enables transient microglia depletion that is followed by microglia repopulation after treatment interruption, causing no known harm to mice. We tested whether this strategy restored microglia function and ameliorated stroke outcome in old mice. Cerebral ischemia/reperfusion induced innate immune responses in microglia highlighted by type I interferon and metabolic changes involving lipid droplet biogenesis. Old microglia accumulated lipids under steady state and displayed exacerbated innate immune responses to stroke. Microglia repopulation in old mice reduced lipid-laden microglia, and the cells exhibited reduced inflammatory responses to ischemia. Moreover, old mice with renewed microglia showed improved motor function 2 weeks after stroke. We conclude that lipid deposits in aged microglia impair the cellular responses to ischemia and worsen functional recovery in old mice.
Collapse
Affiliation(s)
- Maria Arbaizar-Rovirosa
- Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Pedragosa
- Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Juan J Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Carme Casal
- Microscopy Service, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Albert Pol
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Mattia Gallizioli
- Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Anna M Planas
- Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
42
|
Costa MFDS, Pereira-Dutra F, Deboosere N, Jouny S, Song OR, Iack G, Souza AL, Roma EH, Delorme V, Bozza PT, Brodin P. Mycobacterium tuberculosis induces delayed lipid droplet accumulation in dendritic cells depending on bacterial viability and virulence. Mol Microbiol 2023; 119:224-236. [PMID: 36579614 DOI: 10.1111/mmi.15023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 12/30/2022]
Abstract
Tuberculosis remains a global health threat with high morbidity. Dendritic cells (DCs) participate in the acute and chronic inflammatory responses to Mycobacterium tuberculosis (Mtb) by directing the adaptive immune response and are present in lung granulomas. In macrophages, the interaction of lipid droplets (LDs) with mycobacteria-containing phagosomes is central to host-pathogen interactions. However, the data available for DCs are still a matter of debate. Here, we reported that bone marrow-derived DCs (BMDCs) were susceptible to Mtb infection and replication at similar rate to macrophages. Unlike macrophages, the analysis of gene expression showed that Mtb infection induced a delayed increase in lipid droplet-related genes and proinflammatory response. Hence, LD accumulation has been observed by high-content imaging in late periods. Infection of BMDCs with killed H37Rv demonstrated that LD accumulation depends on Mtb viability. Moreover, infection with the attenuated strains H37Ra and Mycobacterium bovis-BCG induced only an early transient increase in LDs, whereas virulent Mtb also induced delayed LD accumulation. In addition, infection with the BCG strain with the reintroduced virulence RD1 locus induced higher LD accumulation and bacterial replication when compared to parental BCG. Collectively, our data suggest that delayed LD accumulation in DCs is dependent on mycobacterial viability and virulence.
Collapse
Affiliation(s)
- Maria Fernanda de Souza Costa
- Instituto de Biologia, Departamento de Imunobiologia, Universidade Federal Fluminense, Niteroi, Brazil.,Center for Technological Development in Health, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Filipe Pereira-Dutra
- Immunopharmacology Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Nathalie Deboosere
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Samuel Jouny
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Ok-Ryul Song
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Guilherme Iack
- Instituto de Biologia, Departamento de Imunobiologia, Universidade Federal Fluminense, Niteroi, Brazil.,Immunopharmacology Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Andreia Lamoglia Souza
- Fundação Oswaldo Cruz, Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Eric Henrique Roma
- Fundação Oswaldo Cruz, Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Vincent Delorme
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Patricia T Bozza
- Immunopharmacology Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Priscille Brodin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
43
|
Birari P, Mal S, Majumder D, Sharma AK, Kumar M, Das T, Ghosh Z, Jana K, Gupta UD, Kundu M, Basu J. Nur77 influences immunometabolism to regulate the release of proinflammatory cytokines and the formation of lipid bodies during Mycobacterium tuberculosis infection of macrophages. Pathog Dis 2023; 81:ftad033. [PMID: 38017622 DOI: 10.1093/femspd/ftad033] [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: 06/12/2023] [Revised: 10/05/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023] Open
Abstract
Infection of macrophages with Mycobacterium tuberculosis induces innate immune responses designed to clear the invading bacterium. However, bacteria often survive within the intracellular environment by exploiting these responses triggered by macrophages. Here, the role of the orphan nuclear receptor Nur77 (Nr4a1) in regulating the response of macrophages infected with M. tuberculosis (Mtb) has been delineated. Nur77 is induced early during infection, regulates metabolism by binding directly at the promoter of the TCA cycle enzyme, isocitrate dehydrogenase 2 (IDH2), to act as its repressor, and shifts the balance from a proinflammatory to an anti-inflammatory phenotype. Depletion of Nur77 increased transcription of IDH2 and, consequently, the levels of intracellular succinate, leading to enhanced levels of the proinflammatory cytokine IL-1β. Further, Nur77 inhibited the production of antibacterial nitric oxide and IL-1β in a succinate dehydrogenase (SDH)-dependent manner, suggesting that its induction favors bacterial survival by suppressing bactericidal responses. Indeed, depletion of Nur77 inhibited the intracellular survival of Mtb. On the other hand, depletion of Nur77 enhanced lipid body formation, suggesting that the fall in Nur77 levels as infection progresses likely favors foamy macrophage formation and long-term survival of Mtb in the host milieu.
Collapse
Affiliation(s)
- Pankaj Birari
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Soumya Mal
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, India
| | - Debayan Majumder
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Arun K Sharma
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Manish Kumar
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Troyee Das
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, India
| | - Zhumur Ghosh
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, India
| | - Kuladip Jana
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, India
| | - Umesh D Gupta
- National JALMA Institute of Leprosy and Other Mycobacterial Disease, Agra 282001, India
| | - Manikuntala Kundu
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Joyoti Basu
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| |
Collapse
|
44
|
de Almeida PE, Pereira de Sousa NM, Rampinelli PG, Silva RVDS, Correa JR, D’Avila H. Lipid droplets as multifunctional organelles related to the mechanism of evasion during mycobacterial infection. Front Cell Infect Microbiol 2023; 13:1102643. [PMID: 36909724 PMCID: PMC9996354 DOI: 10.3389/fcimb.2023.1102643] [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: 11/19/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by the bacteria of the Mycobaterium tuberculosis (Mtb) complex. The modulation of the lipid metabolism has been implicated in the immune response regulation, including the formation of lipid droplets (LD)s, LD-phagosome association and eicosanoid synthesis. Mtb, M. bovis BCG and other pathogenic mycobacteria, as well as wall components, such as LAM, can induce LDs formation in a mechanism involving surface receptors, for instance TLRs, CD36, CD14, CD11b/CD18 and others. In addition, the activation of the lipid-activated nuclear receptor PPARγ is involved in the mechanisms of LD biogenesis, as well as in the modulation of the synthesis of lipid mediators. In infected cells, LDs are sites of compartmentalized prostaglandin E2 synthesis involved in macrophage deactivation, bacterial replication and regulation of the host cytokine profile. LDs also have a function in vesicle traffic during infection. Rab7 and RILP, but not Rab5, are located on LDs of infected macrophages, suggesting that LDs and phagosomes could exchange essential proteins for phagosomal maturation, interfering in mycobacterial survival. The pharmacological inhibition of LDs biogenesis affects the bacterial replication and the synthesis of lipid mediators and cytokines, suggesting that LDs may be new targets for antimicrobial therapies. However, it is still controversial if the accumulation of LDs favors the mycobacterial survival acting as an escape mechanism, or promotes the host resistance to infection. Thus, in this mini-review we discuss recent advances in understanding the important role of LDs in the course of infections and the implications for the pathophysiology of mycobacteriosis.
Collapse
Affiliation(s)
- Patrícia Elaine de Almeida
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
- *Correspondence: Heloisa D’Avila, ; Patrícia Elaine de Almeida, ; José Raimundo Correa,
| | - Núbia Maria Pereira de Sousa
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, University of Brasilia, Brasilia, DF, Brazil
| | - Pollianne Garbero Rampinelli
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | - Renata Vieira de Sousa Silva
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | - José Raimundo Correa
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, University of Brasilia, Brasilia, DF, Brazil
- *Correspondence: Heloisa D’Avila, ; Patrícia Elaine de Almeida, ; José Raimundo Correa,
| | - Heloisa D’Avila
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
- *Correspondence: Heloisa D’Avila, ; Patrícia Elaine de Almeida, ; José Raimundo Correa,
| |
Collapse
|
45
|
Llibre A, Smith N, Rouilly V, Musvosvi M, Nemes E, Posseme C, Mabwe S, Charbit B, Mbandi SK, Filander E, Africa H, Saint-André V, Bondet V, Bost P, Mulenga H, Bilek N, Albert ML, Scriba TJ, Duffy D. Tuberculosis alters immune-metabolic pathways resulting in perturbed IL-1 responses. Front Immunol 2022; 13:897193. [PMID: 36591308 PMCID: PMC9795069 DOI: 10.3389/fimmu.2022.897193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB) remains a major public health problem and we lack a comprehensive understanding of how Mycobacterium tuberculosis (M. tb) infection impacts host immune responses. We compared the induced immune response to TB antigen, BCG and IL-1β stimulation between latently M. tb infected individuals (LTBI) and active TB patients. This revealed distinct responses between TB/LTBI at transcriptomic, proteomic and metabolomic levels. At baseline, we identified a novel immune-metabolic association between pregnane steroids, the PPARγ pathway and elevated plasma IL-1ra in TB. We observed dysregulated IL-1 responses after BCG stimulation in TB patients, with elevated IL-1ra responses being explained by upstream TNF differences. Additionally, distinct secretion of IL-1α/IL-1β in LTBI/TB after BCG stimulation was associated with downstream differences in granzyme mediated cleavage. Finally, IL-1β driven signalling was dramatically perturbed in TB disease but was completely restored after successful treatment. This study improves our knowledge of how immune responses are altered during TB disease, and may support the design of improved preventive and therapeutic tools, including host-directed strategies.
Collapse
Affiliation(s)
- Alba Llibre
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nikaïa Smith
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | | | - Munyaradzi Musvosvi
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Céline Posseme
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Simbarashe Mabwe
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Bruno Charbit
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Stanley Kimbung Mbandi
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Elizabeth Filander
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Hadn Africa
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Violaine Saint-André
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France,Bioinformatics and Biostatistics HUB, Computational Biology Department, Institut Pasteur, Université Paris Cité, Paris, France
| | - Vincent Bondet
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Pierre Bost
- Sorbonne Université, Complexité du vivant, Paris, France,Systems Biology Group, Computational Biology Department, Institut Pasteur, Université Paris Cité, Paris, France
| | - Humphrey Mulenga
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France,Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, Université Paris Cité, Paris, France,*Correspondence: Darragh Duffy,
| |
Collapse
|
46
|
Gong Z, Li Q, Shi J, Liu ET, Shultz LD, Ren G. Lipid-laden lung mesenchymal cells foster breast cancer metastasis via metabolic reprogramming of tumor cells and natural killer cells. Cell Metab 2022; 34:1960-1976.e9. [PMID: 36476935 PMCID: PMC9819197 DOI: 10.1016/j.cmet.2022.11.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/21/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
While the distant organ environment is known to support metastasis of primary tumors, its metabolic roles in this process remain underdetermined. Here, in breast cancer models, we found lung-resident mesenchymal cells (MCs) accumulating neutral lipids at the pre-metastatic stage. This was partially mediated by interleukin-1β (IL-1β)-induced hypoxia-inducible lipid droplet-associated (HILPDA) that subsequently represses adipose triglyceride lipase (ATGL) activity in lung MCs. MC-specific ablation of the ATGL or HILPDA genes in mice reinforced and reduced lung metastasis of breast cancer respectively, suggesting a metastasis-promoting effect of lipid-laden MCs. Mechanistically, lipid-laden MCs transported their lipids to tumor cells and natural killer (NK) cells via exosome-like vesicles, leading to heightened tumor cell survival and proliferation and NK cell dysfunction. Blockage of IL-1β, which was effective singly, improved the efficacy of adoptive NK cell immunotherapy in mitigating lung metastasis. Collectively, lung MCs metabolically regulate tumor cells and anti-tumor immunity to facilitate breast cancer lung metastasis.
Collapse
Affiliation(s)
- Zheng Gong
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Qing Li
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Jiayuan Shi
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Edison T Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | | | - Guangwen Ren
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Tufts University School of Medicine, Boston, MA 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
47
|
Chandra P, Grigsby SJ, Philips JA. Immune evasion and provocation by Mycobacterium tuberculosis. Nat Rev Microbiol 2022; 20:750-766. [PMID: 35879556 PMCID: PMC9310001 DOI: 10.1038/s41579-022-00763-4] [Citation(s) in RCA: 228] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 02/07/2023]
Abstract
Mycobacterium tuberculosis, the causative agent of tuberculosis, has infected humans for millennia. M. tuberculosis is well adapted to establish infection, persist in the face of the host immune response and be transmitted to uninfected individuals. Its ability to complete this infection cycle depends on it both evading and taking advantage of host immune responses. The outcome of M. tuberculosis infection is often a state of equilibrium characterized by immunological control and bacterial persistence. Recent data have highlighted the diverse cell populations that respond to M. tuberculosis infection and the dynamic changes in the cellular and intracellular niches of M. tuberculosis during the course of infection. M. tuberculosis possesses an arsenal of protein and lipid effectors that influence macrophage functions and inflammatory responses; however, our understanding of the role that specific bacterial virulence factors play in the context of diverse cellular reservoirs and distinct infection stages is limited. In this Review, we discuss immune evasion and provocation by M. tuberculosis during its infection cycle and describe how a more detailed molecular understanding is crucial to enable the development of novel host-directed therapies, disease biomarkers and effective vaccines.
Collapse
Affiliation(s)
- Pallavi Chandra
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Steven J Grigsby
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jennifer A Philips
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
48
|
Wan Z, Fu S, Wang Z, Xu Y, Zhou Y, Lin X, Lan R, Han X, Luo Z, Miao J. FABP4-mediated lipid droplet formation in Streptococcus uberis-infected macrophages supports host defence. Vet Res 2022; 53:90. [PMID: 36371263 PMCID: PMC9652580 DOI: 10.1186/s13567-022-01114-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 09/16/2022] [Indexed: 11/15/2022] Open
Abstract
Foamy macrophages containing prominent cytoplasmic lipid droplets (LDs) are found in a variety of infectious diseases. However, their role in Streptococcus uberis-induced mastitis is unknown. Herein, we report that S. uberis infection enhances the fatty acid synthesis pathway in macrophages, resulting in a sharp increase in LD levels, accompanied by a significantly enhanced inflammatory response. This process is mediated by the involvement of fatty acid binding protein 4 (FABP4), a subtype of the fatty acid-binding protein family that plays critical roles in metabolism and inflammation. In addition, FABP4 siRNA inhibitor cell models showed that the deposition of LDs decreased, and the mRNA expression of Tnf, Il1b and Il6 was significantly downregulated after gene silencing. As a result, the bacterial load in macrophages increased. Taken together, these data demonstrate that macrophage LD formation is a host-driven component of the immune response to S. uberis. FABP4 contributes to promoting inflammation via LDs, which should be considered a new target for drug development to treat infections.
Collapse
|
49
|
Singh S, Maurya SK, Aqdas M, Bashir H, Arora A, Bhalla V, Agrewala JN. Mycobacterium tuberculosis exploits MPT64 to generate myeloid-derived suppressor cells to evade the immune system. Cell Mol Life Sci 2022; 79:567. [PMID: 36283989 PMCID: PMC11803053 DOI: 10.1007/s00018-022-04596-5] [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: 06/30/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/24/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is a smart and successful pathogen since it can persist in the intimidating environment of the host by taming and tuning the immune system. Mtb releases MPT64 (Rv1980c) protein in high amounts in patients with active tuberculosis (TB). Consequently, we were curious to decipher the role of MPT64 on the differentiating dendritic cells (DCs) and its relation to evading the immune system. We observed that pre-exposure of differentiating DCs to MPT64 (DCMPT64) transformed them into a phenotype of myeloid-derived suppressor cells (MDSCs). DCMPT64 expressed a high level of immunosuppressive molecules PD-L1, TIM-3, nitric oxide (NO), arginase 1, IDO-1, IL-10 and TGF-β, but inhibited the production of pro-inflammatory cytokines TNF-α, IL-6 and IL-12. DCMPT64 chemotaxis function was diminished due to the reduced expression of CCR7. DCMPT64 promoted the generation of regulatory T cells (Tregs) but inhibited the differentiation of Th1 cells and Th17 cells. Further, high lipid and methylglyoxal content, and reduced glucose consumption by DCMPT64, rendered them metabolically quiescent and consequently, reduced DCMPT64 ability to phagocytose Mtb and provided a safer shelter for the intracellular survival of the mycobacterium. The mechanism identified in impairing the function of DCMPT64 was through the increased production and accumulation of methylglyoxal. Hence, for the first time, we demonstrate the novel role of MPT64 in promoting the generation of MDSCs to favor Mtb survival and escape its destruction by the immune system.
Collapse
Affiliation(s)
- Sanpreet Singh
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Sudeep K Maurya
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Mohammad Aqdas
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Hilal Bashir
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Ashish Arora
- Department of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Vijayender Bhalla
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
- Biosensor Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India.
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, India.
| |
Collapse
|
50
|
Marrocco A, Ortiz LA. Role of metabolic reprogramming in pro-inflammatory cytokine secretion from LPS or silica-activated macrophages. Front Immunol 2022; 13:936167. [PMID: 36341426 PMCID: PMC9633986 DOI: 10.3389/fimmu.2022.936167] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
In the lungs, macrophages constitute the first line of defense against pathogens and foreign bodies and play a fundamental role in maintaining tissue homeostasis. Activated macrophages show altered immunometabolism and metabolic changes governing immune effector mechanisms, such as cytokine secretion characterizing their classic (M1) or alternative (M2) activation. Lipopolysaccharide (LPS)-stimulated macrophages demonstrate enhanced glycolysis, blocked succinate dehydrogenase (SDH), and increased secretion of interleukin-1 beta (IL-1β) and tumor necrosis factor-alpha (TNF-α). Glycolysis suppression using 2 deoxyglucose in LPS-stimulated macrophages inhibits IL-1β secretion, but not TNF-α, indicating metabolic pathway specificity that determines cytokine production. In contrast to LPS, the nature of the immunometabolic responses induced by non-organic particles, such as silica, in macrophages, its contribution to cytokine specification, and disease pathogenesis are not well understood. Silica-stimulated macrophages activate pattern recognition receptors (PRRs) and NLRP3 inflammasome and release IL-1β, TNF-α, and interferons, which are the key mediators of silicosis pathogenesis. In contrast to bacteria, silica particles cannot be degraded, and the persistent macrophage activation results in an increased NADPH oxidase (Phox) activation and mitochondrial reactive oxygen species (ROS) production, ultimately leading to macrophage death and release of silica particles that perpetuate inflammation. In this manuscript, we reviewed the effects of silica on macrophage mitochondrial respiration and central carbon metabolism determining cytokine specification responsible for the sustained inflammatory responses in the lungs.
Collapse
Affiliation(s)
- Antonella Marrocco
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Luis A. Ortiz
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|