1
|
Kim HW, Lee JW, Yoon HS, Park HW, Lee YI, Lee SK, Whang J, Kim JS. Restriction of mitochondrial oxidation of glutamine or fatty acids enhances intracellular growth of Mycobacterium abscessus in macrophages. Virulence 2025; 16:2454323. [PMID: 39828906 PMCID: PMC11749347 DOI: 10.1080/21505594.2025.2454323] [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: 08/18/2024] [Revised: 11/28/2024] [Accepted: 01/12/2025] [Indexed: 01/22/2025] Open
Abstract
Mycobacterium abscessus (Mab), a nontuberculous mycobacterium, is increasing in prevalence worldwide and causes treatment-refractory pulmonary diseases. However, how Mab rewires macrophage energy metabolism to facilitate its survival is poorly understood. We compared the metabolic profiles of murine bone marrow-derived macrophages (BMDMs) infected with smooth (S)- and rough (R)-type Mab using extracellular flux technology. Mab infection shifted BMDMs towards a more energetic phenotype, marked by increased oxidative phosphorylation (OXPHOS) and glycolysis, with a significantly greater enhancement in OXPHOS. This metabolic adaptation was characterized by enhanced ATP production rates, particularly in cells infected with S-type Mab, highlighting OXPHOS as a key energy source. Notably, Mab infection also modulated mitochondrial substrate preferences, increasing fatty acid oxidation capabilities while revealing significant changes in glutamine dependency and flexibility. R-type Mab infections exhibited a marked decrease in glutamine reliance but enhanced metabolic flexibility and capacity. Furthermore, targeting metabolic pathways related to glutamine and fatty acid oxidation exacerbated Mab growth within macrophages, suggesting these pathways play a protective role against infection. These insights advance our understanding of Mab's impact on host cell metabolism and propose a novel avenue for therapeutic intervention. By manipulating host mitochondrial metabolism, we identify a potential host-directed therapeutic strategy against Mab, offering a promising alternative to conventional treatments beleaguered by drug resistance. This study underscores the importance of exploring metabolic interventions to combat Mab infection, paving the way for innovative approaches in the fight against this formidable pathogen.
Collapse
Affiliation(s)
- Ho Won Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Ji Won Lee
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Hoe Sun Yoon
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Hwan-Woo Park
- Department of Cell Biology, Konyang University Hospital and College of Medicine, Daejeon, South Korea
| | | | - Sung Ki Lee
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, South Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC), Department of Research and Development, The Korean Institute of Tuberculosis, Osong, South Korea
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
- Department of Cell Biology, Konyang University Hospital and College of Medicine, Daejeon, South Korea
| |
Collapse
|
2
|
Ashokcoomar S, Naidoo TJ, Cumming BM, Baig Z, Truebody B, Mackenzie JS, Steyn AJC, Pillay M. Mycobacterium tuberculosis curli pili reduces oxygen consumption rate of THP-1 macrophages during early infection. Int J Biochem Cell Biol 2025; 185:106802. [PMID: 40381906 DOI: 10.1016/j.biocel.2025.106802] [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: 01/10/2025] [Revised: 04/25/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
The development of improved anti-tuberculosis (TB) strategies to address drug-resistance and ineffectual TB treatment regimens should focus on interrupting the initial host-pathogen interaction. This study aimed to elucidate the effect of surface-located adhesin, Mycobacterium tuberculosis (Mtb) curli pili (MTP), on the bioenergetic and metabolomic profiles of THP-1 macrophages during initial stages of infection. Differentiated THP-1 macrophages were infected with wildtype (WT), ∆mtp, or mtp-complemented strains of Mtb. Bioenergetic profiles and metabolic flux were determined and statistical analysis highlighted differences/similarities amongst the THP-1 macrophage groups. The ∆mtp infected THP-1 macrophages mimicked the higher oxygen consumption rate (OCR) for basal respiration, ATP production, maximal respiration and spare respiratory capacity of the uninfected THP-1 macrophages, relative to the WT and mtp-complement infected THP-1 macrophages. The ∆mtp infected THP-1 macrophages displayed the highest compensatory glycolytic rate. Mtb infection caused the redirection of carbon from the tricarboxylic acid cycle to glycolysis, in addition to an increased flux through the pentose phosphate pathway. However, in the ∆mtp infected THP-1 macrophages, the total metabolite abundance was lower, similar to the uninfected THP-1 macrophages. Data indicates that the absence of MTP facilitates prompt clearance of the intracellular pathogen before it establishes a successful infection. This implies that the presence of MTP facilitates the survival of the pathogen during the early stages until infection is established. These findings support the growing evidence that the MTP adhesin is an important virulence factor and interruption of the interaction between pathogen and host, will facilitate swift clearance of the infection by the host.
Collapse
Affiliation(s)
- Shinese Ashokcoomar
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st floor Doris Duke Medical Research Institute, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Tarien J Naidoo
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st floor Doris Duke Medical Research Institute, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Bridgette M Cumming
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Zainab Baig
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Barry Truebody
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Jared S Mackenzie
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Adrie J C Steyn
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Manormoney Pillay
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st floor Doris Duke Medical Research Institute, Congella, Private Bag 7, Durban 4013, South Africa.
| |
Collapse
|
3
|
Croock D, Swart Y, Sanko TJ, Mavumengwana V, Möller M, Uren C, Petersen DC. Uncharted territory: the role of mitochondrial DNA variation in macrophage-mediated host susceptibility to tuberculosis. Tuberculosis (Edinb) 2025; 153:102650. [PMID: 40354681 DOI: 10.1016/j.tube.2025.102650] [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: 03/06/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Mitochondria form an integral, yet frequently underappreciated, part of the immune response to Mycobacterium tuberculosis (M.tb), particularly within macrophages. Despite growing recognition for their role in infection and immunity, studies investigating how mitochondrial DNA (mtDNA) variation influences host susceptibility to tuberculosis (TB) are limited. Notably, there are no studies in African-based populations, although Africans possess unparalleled human genetic diversity, including the earliest diverged mitochondrial haplogroups, and a high TB burden. This underrepresentation limits the discovery of novel ancestry-specific genetic loci associated with TB. In this review article, we describe the unique characteristics of mtDNA, highlight key mitochondrial functions relevant to macrophage responses during M.tb infection, and summarise published studies that investigate the role of host mtDNA variation in TB susceptibility. We further advocate for the inclusion of African populations in future studies to identify novel TB susceptibility genetic risk loci and expand the current knowledgebase on host TB susceptibility.
Collapse
Affiliation(s)
- Dayna Croock
- South African Medical Research Council (SAMRC) Centre for Tuberculosis Research (CTR), Division of Molecular Biology and Human Genetics (MBHG), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Yolandi Swart
- South African Medical Research Council (SAMRC) Centre for Tuberculosis Research (CTR), Division of Molecular Biology and Human Genetics (MBHG), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Tomasz Janusz Sanko
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - Vuyo Mavumengwana
- South African Medical Research Council (SAMRC) Centre for Tuberculosis Research (CTR), Division of Molecular Biology and Human Genetics (MBHG), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Marlo Möller
- South African Medical Research Council (SAMRC) Centre for Tuberculosis Research (CTR), Division of Molecular Biology and Human Genetics (MBHG), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; Centre for Bioinformatics and Computational Biology, Stellenbosch University, Cape Town, South Africa; Genomics for Health in Africa (GHA), Africa-Europe Cluster of Research Excellence (CoRE), South Africa; National Institute for Theoretical and Computational Sciences (NITheCS) South Africa, South Africa
| | - Caitlin Uren
- South African Medical Research Council (SAMRC) Centre for Tuberculosis Research (CTR), Division of Molecular Biology and Human Genetics (MBHG), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; Centre for Bioinformatics and Computational Biology, Stellenbosch University, Cape Town, South Africa; Genomics for Health in Africa (GHA), Africa-Europe Cluster of Research Excellence (CoRE), South Africa; National Institute for Theoretical and Computational Sciences (NITheCS) South Africa, South Africa
| | - Desiree C Petersen
- South African Medical Research Council (SAMRC) Centre for Tuberculosis Research (CTR), Division of Molecular Biology and Human Genetics (MBHG), Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; Genomics for Health in Africa (GHA), Africa-Europe Cluster of Research Excellence (CoRE), South Africa.
| |
Collapse
|
4
|
Brown K, Walsh A, Yennemadi A, O'Leary S, O'Sullivan M, Nadarajan P, Basdeo S, Leisching G, Keane J. Mycobacterium tuberculosis Induces Warburg Metabolism in Human Alveolar Macrophages: A Transcriptomic Analysis. Am J Respir Cell Mol Biol 2025; 72:597-602. [PMID: 40311077 DOI: 10.1165/rcmb.2024-0268le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
Affiliation(s)
- Kevin Brown
- Trinity College Dublin, Ireland
- Saint James' Hospital Dublin, Ireland
| | - Aaron Walsh
- Trinity College Dublin, Ireland
- Saint James' Hospital Dublin, Ireland
| | - Anjali Yennemadi
- Trinity College Dublin, Ireland
- Saint James' Hospital Dublin, Ireland
| | - Seónadh O'Leary
- Trinity College Dublin, Ireland
- Saint James' Hospital Dublin, Ireland
| | - Mary O'Sullivan
- Trinity College Dublin, Ireland
- Saint James' Hospital Dublin, Ireland
| | | | - Sharee Basdeo
- Trinity College Dublin, Ireland
- Saint James' Hospital Dublin, Ireland
| | - Gina Leisching
- Trinity College Dublin, Ireland
- Saint James' Hospital Dublin, Ireland
| | - Joseph Keane
- Trinity College Dublin, Ireland
- Saint James' Hospital Dublin, Ireland
| |
Collapse
|
5
|
Fernandes AI, Pinto AJ, Silvério D, Zedler U, Ferreira C, Duarte IF, Silvestre R, Dorhoi A, Saraiva M. Genetically Diverse Mycobacterium tuberculosis Isolates Manipulate Inflammasome and Interleukin 1β Secretion Independently of Macrophage Metabolic Rewiring. J Infect Dis 2025; 231:e671-e684. [PMID: 39570738 PMCID: PMC11998582 DOI: 10.1093/infdis/jiae583] [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: 06/25/2024] [Accepted: 11/19/2024] [Indexed: 04/17/2025] Open
Abstract
The diversity of Mycobacterium tuberculosis impacts the outcome of tuberculosis. We previously showed that M. tuberculosis isolates obtained from patients with severe disease induced low inflammasome activation and interleukin 1β (IL-1β) production by infected macrophages. Here we questioned whether this differential modulation of macrophages by M. tuberculosis isolates depended on distinct metabolic reprogramming. We found that the macrophage metabolic landscape was similar regardless of the infecting M. tuberculosis isolate. Paralleling single-Toll-like receptor (TLR) activated macrophages, glycolysis inhibition during infection impaired IL-1β secretion. However, departing from TLR -based models, in infected macrophages, IL-1β secretion was independent of mitochondrial metabolic changes and hypoxia-inducible factor 1α (HIF-1α). Additionally, we found an unappreciated impact of a host metabolic inhibitor on the pathogen, and show that inflammasome activation and IL-1β production by macrophages require metabolically active bacteria. Our study highlights the potential confounding effect of host metabolic inhibitors on the pathogen and uncoupling of M. tuberculosis-inflammasome modulation from the host metabolic reprogramming.
Collapse
Affiliation(s)
- Ana Isabel Fernandes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Doctoral Program in Molecular and Cell Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Alexandre Jorge Pinto
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Diogo Silvério
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Doctoral Program in Molecular and Cell Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ulrike Zedler
- Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Carolina Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Iola F Duarte
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| | - Margarida Saraiva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| |
Collapse
|
6
|
Alhendi A, Naser SA. MCT4 inhibition attenuates inflammatory response to Mycobacterium avium paratuberculosis infection and restores intestinal epithelial integrity in vitro. Front Immunol 2025; 16:1562100. [PMID: 40297589 PMCID: PMC12034541 DOI: 10.3389/fimmu.2025.1562100] [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: 01/16/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Mycobacterium avium paratuberculosis (MAP) plays a significant role in Crohn's disease (CD). Monocarboxylate transporter 4 (MCT4) is a proton-coupled symporter of lactate that facilitates the inflammatory shift in macrophages and increases their reliance on glycolysis. MCT4 is also involved in the negative regulation of intestinal epithelial barrier function. Methods In this in vitro study, we examined the role of MCT4 in macrophages and its effect on intestinal epithelial homeostasis during MAP infection. We used cultured THP-1 macrophages infected with a clinical strain of MAP (UCF4) as well as intestinal cell lines, Caco-2 and HT-29. MCT4 was inhibited using α-cyano-4-hydroxycinnamic acid (CHCα). Results Infection of THP-1 cells with MAP upregulated MCT4 expression (2 folds) and resulted in a significant increase in lactate export (1.3 folds), TNFα (13.8 folds), and IL-6 (1.3) via TLR2 activation. Consequently, intestinal damage markers were also upregulated, including MUC2 (2.5 folds), NOX-1 (2 folds), SERPINE1 (2.1 folds), IL-6 (1.6 folds), and CLDN2 (1.4 folds). Inhibition of MCT4 during MAP infection with CHCα significantly reduced TNF-α and IL-6 levels. This effect on macrophages restored baseline oxidative status and mucin production in HT-29 intestinal cells. Moreover, MCT4 inhibition in a MAP-infected THP-1-Caco-2 co-culture system restored IL-6 and SERPINE1 to normal levels and enhanced tight junction protein, TJP1 (ZO-1), expression. Conclusion Collectively, this study revealed the significant role of MCT4 in CD pathophysiology during MAP infection and highlighted MCT4 as a potential therapeutic target for CD treatment.
Collapse
Affiliation(s)
| | - Saleh A. Naser
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
7
|
Hajra D, Yadav V, Singh A, Chakravortty D. SIRT1 and SIRT3 Impact Host Mitochondrial Function and Host Salmonella pH Balance during Infection. ACS Infect Dis 2025; 11:827-843. [PMID: 40168249 DOI: 10.1021/acsinfecdis.4c00751] [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: 04/03/2025]
Abstract
Mitochondria are important organelles that regulate energy homeostasis. Mitochondrial health and dynamics are crucial determinants of the outcome of several bacterial infections. SIRT3, a major mitochondrial sirtuin, along with SIRT1 regulates key mitochondrial functions. This led to considerable interest in understanding the role of SIRT1 and SIRT3 in governing mitochondrial functions during Salmonella infection. Here, we show that loss of SIRT1 and SIRT3 function either by shRNA-mediated knockdown or by inhibitor treatment led to increased mitochondrial dysfunction with alteration in mitochondrial bioenergetics alongside increased mitochondrial superoxide generation in Salmonella-infected macrophages. Consistent with dysfunctional mitochondria, mitophagy was induced along with altered mitochondrial fusion-fission dynamics in S. typhimurium-infected macrophages. Additionally, the mitochondrial bioenergetic alteration promotes acidification of the infected macrophage cytosolic pH. This host cytosolic pH imbalance skewed the intraphagosomal and intrabacterial pH in the absence of SIRT1 and SIRT3, resulting in decreased SPI-2 gene expression. Our results suggest a novel role for SIRT1 and SIRT3 in maintaining the intracellular Salmonella niche by modulating the mitochondrial bioenergetics and dynamics in the infected macrophages.
Collapse
Affiliation(s)
- Dipasree Hajra
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Vikas Yadav
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Amit Singh
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Dipshikha Chakravortty
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, India
- Adjunct Faculty, School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala 695551, India
| |
Collapse
|
8
|
Lohia GK, Riquelme SA. Influence of cell bioenergetics on host-pathogen interaction in the lung. Front Immunol 2025; 16:1549293. [PMID: 40248701 PMCID: PMC12003392 DOI: 10.3389/fimmu.2025.1549293] [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: 12/20/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Pulmonary diseases, arising from infections caused by bacteria, fungi, and viruses, or stemming from underlying genetic factors are one of the leading causes of mortality in humans, accounting for millions of deaths every year. At the onset of pulmonary diseases, crucial roles are played by phagocytic immune cells, particularly tissue-resident macrophages, in regulating the immune response at the mucosal barrier. Recent strides have illuminated the pivotal role of host bioenergetics modulated by metabolites derived from both pathogens and hosts in influencing the pathophysiology of major organs. Their influence extends to processes such as the infiltration of immune cells, activation of macrophages, and the polarization phenomenon. Furthermore, host-derived metabolites, such as itaconate, contribute to the promotion of anti-inflammatory responses, thereby preventing immunopathology and facilitating the preservation of mucosal niches to thrive for the long-term. This review explores recent advancements in the field of immunometabolism, with a particular emphasis on the intricacies of disease progression in pulmonary infections caused by bacteria such as P. aeruginosa, M. tuberculosis and S. aureus and fungi like C. albicans.
Collapse
|
9
|
Krueger G, Faisal S, Dorhoi A. Microenvironments of tuberculous granuloma: advances and opportunities for therapy. Front Immunol 2025; 16:1575133. [PMID: 40196129 PMCID: PMC11973276 DOI: 10.3389/fimmu.2025.1575133] [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: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
The hallmark tissue lesions of tuberculosis (TB) are granulomas. These multicellular structures exhibit varying degrees of cellular complexity, are dynamic, and show considerable diversity within and between hosts. Categorization based on gross pathologic features, particularly caseation and necrosis, was historically coined prior to the identification of mycobacteria as the causative agent of TB. More recently, granuloma zonation based on immune cell composition, metabolite abundance, and physical characteristics has gained attention. With the advent of single-cell analyses, distinct microenvironments and cellular ecosystems within TB granulomas have been identified. We summarize the architecture of TB granulomas and highlight their cellular heterogeneity, including cell niches as well as physical factors such as oxygen gradients that modulate lesion fate. We discuss opportunities for therapy, highlighting new models and the power of in silico modeling to unravel granuloma features and trajectories. Understanding the relevance of the granuloma microenvironment to disease pathophysiology will facilitate the development of more effective interventions, such as host-directed therapies for TB.
Collapse
Affiliation(s)
- Gesa Krueger
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Shah Faisal
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
10
|
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
|
11
|
党 怡, 孙 鑫, 蒋 静. [Role of Traditional Chinese Medicine in Pathogen-Induced Metabolic Reprogramming and Immune Suppression]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2025; 56:10-18. [PMID: 40109470 PMCID: PMC11914019 DOI: 10.12182/20250160501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Indexed: 03/22/2025]
Abstract
Traditional Chinese medicine (TCM), an essential component of the traditional medicine practiced in China, has demonstrated unique therapeutic efficacy in combating infectious diseases caused by pathogens and various types of tumors. In particular, TCM plays a vital role in enhancing immune function, maintaining homeostasis, and improving metabolic balance. However, the complex ingredients used in TCM and its broad range of therapeutic targets present challenges for comprehensive analysis of the mechanisms involved. Moreover, the molecular and cellular mechanisms underlying TCM's effects remain underexplored, limiting its broader application in modern medicine. Recent studies have increasingly revealed that TCM can not only directly inhibit the activity of pathogens, such as bacteria, fungi, viruses, and tumor cells, but also exert profound effects on immune remodeling by regulating the metabolism of both pathogens and hosts. Therefore, a comprehensive understanding of the role of TCM in metabolic regulation is crucial for elucidating its anti-pathogenic effects. This review is focused on the metabolic pathways of pathogens and host metabolic reprogramming induced by pathogens. We systematically reviewed the mechanisms by which TCM regulates pathogen metabolism, influences pathogen-induced metabolic reprogramming in hosts, and mitigates immune suppression caused by pathogens. This review may provide new ideas for investigating the molecular mechanisms of TCM in disease development and progression.
Collapse
Affiliation(s)
- 怡雄 党
- 四川大学华西公共卫生学院/四川大学华西第四医院 (成都 610041)West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - 鑫 孙
- 四川大学华西公共卫生学院/四川大学华西第四医院 (成都 610041)West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 中国循证医学中心 (成都 610041)Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 静文 蒋
- 四川大学华西公共卫生学院/四川大学华西第四医院 (成都 610041)West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
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
|
13
|
Lyu J, Narum DE, Baldwin SL, Larsen SE, Bai X, Griffith DE, Dartois V, Naidoo T, Steyn AJC, Coler RN, Chan ED. Understanding the development of tuberculous granulomas: insights into host protection and pathogenesis, a review in humans and animals. Front Immunol 2024; 15:1427559. [PMID: 39717773 PMCID: PMC11663721 DOI: 10.3389/fimmu.2024.1427559] [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: 05/04/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Granulomas, organized aggregates of immune cells which form in response to Mycobacterium tuberculosis (Mtb), are characteristic but not exclusive of tuberculosis (TB). Despite existing investigations on TB granulomas, the determinants that differentiate host-protective granulomas from granulomas that contribute to TB pathogenesis are often disputed. Thus, the goal of this narrative review is to help clarify the existing literature on such determinants. We adopt the a priori view that TB granulomas are host-protective organelles and discuss the molecular and cellular determinants that induce protective granulomas and those that promote their failure. While reports about protective TB granulomas and their failure may initially seem contradictory, it is increasingly recognized that either deficiencies or excesses of the molecular and cellular components in TB granuloma formation may be detrimental to the host. More specifically, insufficient or excessive expression/representation of the following components have been reported to skew granulomas toward the less protective phenotype: (i) epithelioid macrophages; (ii) type 1 adaptive immune response; (iii) type 2 adaptive immune response; (iv) tumor necrosis factor; (v) interleukin-12; (vi) interleukin-17; (vii) matrix metalloproteinases; (viii) hypoxia in the TB granulomas; (ix) hypoxia inducible factor-1 alpha; (x) aerobic glycolysis; (xi) indoleamine 2,3-dioxygenase activity; (xii) heme oxygenase-1 activity; (xiii) immune checkpoint; (xiv) leukotriene A4 hydrolase activity; (xv) nuclear-factor-kappa B; and (xvi) transforming growth factor-beta. Rather, more precise and timely coordinated immune responses appear essential for eradication or containment of Mtb infection. Since there are several animal models of infection with Mtb, other species within the Mtb complex, and the surrogate Mycobacterium marinum - whether natural (cattle, elephants) or experimental (zebrafish, mouse, guinea pig, rabbit, mini pig, goat, non-human primate) infections - we also compared the TB granulomatous response and other pathologic lung lesions in various animals infected with one of these mycobacteria with that of human pulmonary TB. Identifying components that dictate the formation of host-protective granulomas and the circumstances that result in their failure can enhance our understanding of the macrocosm of human TB and facilitate the development of novel remedies - whether they be direct therapeutics or indirect interventions - to efficiently eliminate Mtb infection and prevent its pathologic sequelae.
Collapse
Affiliation(s)
- Jiwon Lyu
- Division of Pulmonary and Critical Medicine, Soon Chun Hyang University Cheonan Hospital, Seoul, Republic of Korea
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Drew E. Narum
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Susan L. Baldwin
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Sasha E. Larsen
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Xiyuan Bai
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - David E. Griffith
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Threnesan Naidoo
- Departments of Forensic & Legal Medicine and Laboratory Medicine & Pathology, Faculty of Medicine & Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Adrie J. C. Steyn
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology and Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rhea N. Coler
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Edward D. Chan
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
14
|
Sanchez SE, Chiarelli TJ, Park MA, Carlyon JA. Orientia tsutsugamushi infection reduces host gluconeogenic but not glycolytic substrates. Infect Immun 2024; 92:e0028424. [PMID: 39324805 PMCID: PMC11556148 DOI: 10.1128/iai.00284-24] [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: 07/13/2024] [Accepted: 08/20/2024] [Indexed: 09/27/2024] Open
Abstract
Orientia tsutsugamushi a causal agent of scrub typhus, is an obligate intracellular bacterium that, akin to other rickettsiae, is dependent on host cell-derived nutrients for survival and thus pathogenesis. Based on limited experimental evidence and genome-based in silico predictions, O. tsutsugamushi is hypothesized to parasitize host central carbon metabolism (CCM). Here, we (re-)evaluated O. tsutsugamushi dependency on host cell CCM as initiated by glucose and glutamine. Orientia infection had no effect on host glucose and glutamine consumption or lactate accumulation, indicating no change in overall flux through CCM. However, host cell mitochondrial activity and ATP levels were reduced during infection and correspond with lower intracellular glutamine and glutamate pools. To further probe the essentiality of host CCM in O. tsutsugamushi proliferation, we developed a minimal medium for host cell cultivation and paired it with chemical inhibitors to restrict the intermediates and processes related to glucose and glutamine metabolism. These conditions failed to negatively impact O. tsutsugamushi intracellular growth, suggesting the bacterium is adept at scavenging from host CCM. Accordingly, untargeted metabolomics was utilized to evaluate minor changes in host CCM metabolic intermediates across O. tsutsugamushi infection and revealed that pathogen proliferation corresponds with reductions in critical CCM building blocks, including amino acids and TCA cycle intermediates, as well as increases in lipid catabolism. This study directly correlates O. tsutsugamushi proliferation to alterations in host CCM and identifies metabolic intermediates that are likely critical for pathogen fitness.IMPORTANCEObligate intracellular bacterial pathogens have evolved strategies to reside and proliferate within the eukaryotic intracellular environment. At the crux of this parasitism is the balance between host and pathogen metabolic requirements. The physiological basis driving O. tsutsugamushi dependency on its mammalian host remains undefined. By evaluating alterations in host metabolism during O. tsutsugamushi proliferation, we discovered that bacterial growth is independent of the host's nutritional environment but appears dependent on host gluconeogenic substrates, including amino acids. Given that O. tsutsugamushi replication is essential for its virulence, this study provides experimental evidence for the first time in the post-genomic era of metabolic intermediates potentially parasitized by a scrub typhus agent.
Collapse
Affiliation(s)
- Savannah E. Sanchez
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Travis J. Chiarelli
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Margaret A. Park
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
15
|
Mal S, Majumder D, Birari P, Sharma AK, Gupta U, Jana K, Kundu M, Basu J. The miR-26a/SIRT6/HIF-1α axis regulates glycolysis and inflammatory responses in host macrophages during Mycobacterium tuberculosis infection. FEBS Lett 2024; 598:2592-2614. [PMID: 39155147 DOI: 10.1002/1873-3468.15001] [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: 03/03/2024] [Revised: 06/12/2024] [Accepted: 07/03/2024] [Indexed: 08/20/2024]
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis. Here, a macrophage infection model was used to unravel the role of the histone deacetylase sirtuin 6 (SIRT6) in Mtb-triggered regulation of the innate immune response. Mtb infection downregulated microRNA-26a and upregulated its target SIRT6. SIRT6 suppressed glycolysis and expression of HIF-1α-dependent glycolytic genes during infection. In addition, SIRT6 regulated the levels of intracellular succinate which controls stabilization of HIF-1α, as well as the release of interleukin (IL)-1β. Furthermore, SIRT6 inhibited inducible nitric oxide synthase (iNOS) and proinflammatory IL-6 but augmented anti-inflammatory arginase expression. The miR-26a/SIRT6/HIF-1α axis therefore regulates glycolysis and macrophage immune responses during Mtb infection. Our findings link SIRT6 to rewiring of macrophage signaling pathways facilitating dampening of the antibacterial immune response.
Collapse
Affiliation(s)
- Soumya Mal
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, Kolkata, India
| | | | - Pankaj Birari
- Department of Chemical Sciences, Bose Institute, Kolkata, India
| | | | - Umesh Gupta
- National JALMA Institute of Leprosy and Other Mycobacterial Disease, Agra, India
| | - Kuladip Jana
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, Kolkata, India
| | | | - Joyoti Basu
- Department of Chemical Sciences, Bose Institute, Kolkata, India
| |
Collapse
|
16
|
Chakraborty A, Bandyopadhaya A, Singh VK, Kovacic F, Cha S, Oldham WM, Tzika AA, Rahme LG. The bacterial quorum sensing signal 2'-aminoacetophenone rewires immune cell bioenergetics through the Ppargc1a/Esrra axis to mediate tolerance to infection. eLife 2024; 13:RP97568. [PMID: 39269443 PMCID: PMC11398867 DOI: 10.7554/elife.97568] [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] [Indexed: 09/15/2024] Open
Abstract
How bacterial pathogens exploit host metabolism to promote immune tolerance and persist in infected hosts remains elusive. To achieve this, we show that Pseudomonas aeruginosa (PA), a recalcitrant pathogen, utilizes the quorum sensing (QS) signal 2'-aminoacetophenone (2-AA). Here, we unveil how 2-AA-driven immune tolerization causes distinct metabolic perturbations in murine macrophages' mitochondrial respiration and bioenergetics. We present evidence indicating that these effects stem from decreased pyruvate transport into mitochondria. This reduction is attributed to decreased expression of the mitochondrial pyruvate carrier (Mpc1), which is mediated by diminished expression and nuclear presence of its transcriptional regulator, estrogen-related nuclear receptor alpha (Esrra). Consequently, Esrra exhibits weakened binding to the Mpc1 promoter. This outcome arises from the impaired interaction between Esrra and the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Ppargc1a). Ultimately, this cascade results in diminished pyruvate influx into mitochondria and, consequently reduced ATP production in tolerized murine and human macrophages. Exogenously added ATP in infected macrophages restores the transcript levels of Mpc1 and Esrra and enhances cytokine production and intracellular bacterial clearance. Consistent with the in vitro findings, murine infection studies corroborate the 2-AA-mediated long-lasting decrease in ATP and acetyl-CoA and its association with PA persistence, further supporting this QS signaling molecule as the culprit of the host bioenergetic alterations and PA persistence. These findings unveil 2-AA as a modulator of cellular immunometabolism and reveal an unprecedented mechanism of host tolerance to infection involving the Ppargc1a/Esrra axis in its influence on Mpc1/OXPHOS-dependent energy production and PA clearance. These paradigmatic findings pave the way for developing treatments to bolster host resilience to pathogen-induced damage. Given that QS is a common characteristic of prokaryotes, it is likely that 2-AA-like molecules with similar functions may be present in other pathogens.
Collapse
Affiliation(s)
- Arijit Chakraborty
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
| | - Arunava Bandyopadhaya
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
| | - Vijay K Singh
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
| | - Filip Kovacic
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
- Institute of Molecular Enzyme Technology, Heinrich Heine University DüsseldorfJülichGermany
| | - Sujin Cha
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
| | - William M Oldham
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - A Aria Tzika
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
| | - Laurence G Rahme
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
17
|
Kumar R, Kolloli A, Subbian S, Kaushal D, Shi L, Tyagi S. Imaging the Architecture of Granulomas Induced by Mycobacterium tuberculosis Infection with Single-molecule Fluorescence In Situ Hybridization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:526-537. [PMID: 38912840 PMCID: PMC11407750 DOI: 10.4049/jimmunol.2300068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/30/2024] [Indexed: 06/25/2024]
Abstract
Granulomas are an important hallmark of Mycobacterium tuberculosis infection. They are organized and dynamic structures created when immune cells assemble around the sites of infection in the lungs that locally restrict M. tuberculosis growth and the host's inflammatory responses. The cellular architecture of granulomas is traditionally studied by immunofluorescence labeling of surface markers on the host cells. However, very few Abs are available for model animals used in tuberculosis research, such as nonhuman primates and rabbits, and secreted immunological markers such as cytokines cannot be imaged in situ using Abs. Furthermore, traditional phenotypic surface markers do not provide sufficient resolution for the detection of the many subtypes and differentiation states of immune cells. Using single-molecule fluorescence in situ hybridization (smFISH) and its derivatives, amplified smFISH and iterative smFISH, we developed a platform for imaging mRNAs encoding immune markers in rabbit and macaque tuberculosis granulomas. Multiplexed imaging for several mRNA and protein markers was followed by quantitative measurement of the expression of these markers in single cells. An analysis of the combinatorial expressions of these markers allowed us to classify the cells into several subtypes, and to chart their densities within granulomas. For one mRNA target, hypoxia-inducible factor-1α, we imaged its mRNA and protein in the same cells, demonstrating the specificity of the probes. This method paves the way for defining granular differentiation states and cell subtypes from transcriptomic data, identifying key mRNA markers for these cell subtypes, and then locating the cells in the spatial context of granulomas.
Collapse
Affiliation(s)
- Ranjeet Kumar
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Afsal Kolloli
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Lanbo Shi
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Sanjay Tyagi
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
18
|
Dias HF, Fu JF, Luck TG, Wolfe GE, Hostetter ER, Ng NC, Zheng H, Kühtreiber WM, Price JC, Catana C, Faustman DL. The spleen assumes a major role in blood glucose regulation in type 1 diabetes patients treated with BCG. Sci Rep 2024; 14:17611. [PMID: 39080423 PMCID: PMC11289084 DOI: 10.1038/s41598-024-67905-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: 02/20/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024] Open
Abstract
The Bacillus Calmette-Guérin (BCG) vaccine, which has been used for > 100 years to prevent tuberculosis, is well-established for bladder cancer treatment, and under study for neurological and autoimmune diseases. In patients with type 1 diabetes (T1D), BCG vaccinations have been shown in randomized clinical trials to gradually lower blood sugar to near normal levels. This effect appears to be driven by a BCG-induced shift in lymphoid cells' glucose metabolism from oxidative phosphorylation to aerobic glycolysis. The latter is a state of high glucose utilization that draws more glucose from the blood. Apart from blood, it is unknown whether BCG establishes residence in any organs and alters their glucose metabolism. In this two-year-long clinical trial in type 1 diabetics, we use positron emission tomography (PET) and x-ray computed tomography (CT) to map organs that increase their uptake of the glucose analogue 18F-fluorodeoxyglucose (18F-FDG) before versus after BCG vaccinations. We also injected BALB/c mice with BCG to test for the presence of BCG in various organs. Results from both studies point to the spleen as the dominant site for glucose uptake and BCG residence. The human spleen is significant because its 47% increase in 18F-FDG uptake by a large population of lymphocytes and monocytes might help to explain BCG's systemic lowering of blood glucose to near normal levels. Findings suggest that the spleen, triggered by BCG, assumes a critical role in systemic glucose regulation in the absence of a functional pancreas.
Collapse
Affiliation(s)
- Hans F Dias
- Massachusetts General Hospital, Boston, MA, 02116, USA
| | - Jessie Fanglu Fu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02116, USA
| | - Trevor G Luck
- Massachusetts General Hospital, Boston, MA, 02116, USA
| | - Grace E Wolfe
- Massachusetts General Hospital, Boston, MA, 02116, USA
| | | | - Nathan C Ng
- Massachusetts General Hospital, Boston, MA, 02116, USA
| | - Hui Zheng
- Statistics Department, Massachusetts General Hospital, Boston, MA, 02116, USA
| | - Willem M Kühtreiber
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, 02116, USA
| | - Julie C Price
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02116, USA
| | - Ciprian Catana
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02116, USA
| | - Denise L Faustman
- Massachusetts General Hospital, Boston, MA, 02116, USA.
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, 02116, USA.
| |
Collapse
|
19
|
Chakraborty A, Bandyopadhaya A, Singh V, Kovacic F, Cha S, Oldham W, Tzika AA, Rahme L. The Bacterial Quorum-Sensing Signal 2-Aminoacetophenone Rewires Immune Cell Bioenergetics through the PGC-1α/ERRα Axis to Mediate Tolerance to Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582124. [PMID: 38464050 PMCID: PMC10925214 DOI: 10.1101/2024.02.26.582124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
How bacterial pathogens exploit host metabolism to promote immune tolerance and persist in infected hosts remains elusive. To achieve this, we show that Pseudomonas aeruginosa (PA), a recalcitrant pathogen, utilizes the quorum sensing (QS) signal 2-aminoacetophenone (2-AA). Here, we unveil how 2-AA-driven immune tolerization causes distinct metabolic perturbations in macrophages mitochondrial respiration and bioenergetics. We present evidence indicating that these effects stem from decreased pyruvate transport into mitochondria. This reduction is attributed to decreased expression of the mitochondrial pyruvate carrier (MPC1), which is mediated by diminished expression and nuclear presence of its transcriptional regulator, estrogen-related nuclear receptor alpha (ERRα). Consequently, ERRα exhibits weakened binding to the MPC1 promoter. This outcome arises from the impaired interaction between ERRα and the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Ultimately, this cascade results in diminished pyruvate influx into mitochondria and, consequently reduced ATP production in tolerized macrophages. Exogenously added ATP in infected macrophages restores the transcript levels of MPC1 and ERRα and enhances cytokine production and intracellular bacterial clearance. Consistent with the in vitro findings, murine infection studies corroborate the 2-AA-mediated long-lasting decrease in ATP and acetyl-CoA and its association with PA persistence, further supporting this QS signaling molecule as the culprit of the host bioenergetic alterations and PA persistence. These findings unveil 2-AA as a modulator of cellular immunometabolism and reveal an unprecedented mechanism of host tolerance to infection involving the PGC-1α/ERRα axis in its influence on MPC1/OXPHOS-dependent energy production and PA clearance. These paradigmatic findings pave the way for developing treatments to bolster host resilience to pathogen-induced damage. Given that QS is a common characteristic of prokaryotes, it is likely that 2-AA-like molecules with similar functions may be present in other pathogens.
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
|
Chen Z, Kong X, Ma Q, Chen J, Zeng Y, Liu H, Wang X, Lu S. The impact of Mycobacterium tuberculosis on the macrophage cholesterol metabolism pathway. Front Immunol 2024; 15:1402024. [PMID: 38873598 PMCID: PMC11169584 DOI: 10.3389/fimmu.2024.1402024] [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: 03/16/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an intracellular pathogen capable of adapting and surviving within macrophages, utilizing host nutrients for its growth and replication. Cholesterol is the main carbon source during the infection process of Mtb. Cholesterol metabolism in macrophages is tightly associated with cell functions such as phagocytosis of pathogens, antigen presentation, inflammatory responses, and tissue repair. Research has shown that Mtb infection increases the uptake of low-density lipoprotein (LDL) and cholesterol by macrophages, and enhances de novo cholesterol synthesis in macrophages. Excessive cholesterol is converted into cholesterol esters, while the degradation of cholesterol esters in macrophages is inhibited by Mtb. Furthermore, Mtb infection suppresses the expression of ATP-binding cassette (ABC) transporters in macrophages, impeding cholesterol efflux. These alterations result in the massive accumulation of cholesterol in macrophages, promoting the formation of lipid droplets and foam cells, which ultimately facilitates the persistent survival of Mtb and the progression of tuberculosis (TB), including granuloma formation, tissue cavitation, and systemic dissemination. Mtb infection may also promote the conversion of cholesterol into oxidized cholesterol within macrophages, with the oxidized cholesterol exhibiting anti-Mtb activity. Recent drug development has discovered that reducing cholesterol levels in macrophages can inhibit the invasion of Mtb into macrophages and increase the permeability of anti-tuberculosis drugs. The development of drugs targeting cholesterol metabolic pathways in macrophages, as well as the modification of existing drugs, holds promise for the development of more efficient anti-tuberculosis medications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaomin Wang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Shuihua Lu
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
22
|
Kyung Kim J, Jo EK. Host and microbial regulation of mitochondrial reactive oxygen species during mycobacterial infections. Mitochondrion 2024; 75:101852. [PMID: 38360196 DOI: 10.1016/j.mito.2024.101852] [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: 10/16/2023] [Revised: 01/15/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
Mycobacteria, including Mycobacterium tuberculosis (Mtb) and non-tuberculous mycobacteria (NTM), pose challenges in treatment due to their increased resistance to antibiotics. Following infection, mycobacteria and their components trigger robust innate and inflammatory immune responses intricately associated with the modulation of mitochondrial functions, including oxidative phosphorylation (OXPHOS) and metabolism. Certainly, mitochondrial reactive oxygen species (mtROS) are an inevitable by-product of OXPHOS and function as a bactericidal weapon; however, an excessive accumulation of mtROS are linked to pathological inflammation and necroptotic cell death during mycobacterial infection. Despite previous studies outlining various host pathways involved in regulating mtROS levels during antimicrobial responses in mycobacterial infection, our understanding of the precise mechanisms orchestrating the fine regulation of this response remains limited. Emerging evidence suggests that mycobacterial proteins play a role in targeting the mitochondria of the host, indicating the potential influence of microbial factors on mitochondrial functions within host cells. In this review, we provide an overview of how both host and Mtb factors influence mtROS generation during infection. A comprehensive study of host and microbial factors that target mtROS will shed light on innovative approaches for effectively managing drug-resistant mycobacterial infections.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Eun-Kyeong Jo
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
23
|
Yang N, Sun M, Wang H, Hu D, Zhang A, Khan S, Chen Z, Chen D, Xie S. Progress of stimulus responsive nanosystems for targeting treatment of bacterial infectious diseases. Adv Colloid Interface Sci 2024; 324:103078. [PMID: 38215562 DOI: 10.1016/j.cis.2024.103078] [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: 08/17/2023] [Revised: 12/27/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
In recent decades, due to insufficient concentration at the lesion site, low bioavailability and increasingly serious resistance, antibiotics have become less and less dominant in the treatment of bacterial infectious diseases. It promotes the development of efficient drug delivery systems, and is expected to achieve high absorption, targeted drug release and satisfactory therapy effects. A variety of endogenous stimulation-responsive nanosystems have been constructed by using special infection microenvironments (pH, enzymes, temperature, etc.). In this review, we firstly provide an extensive review of the current research progress in antibiotic treatment dilemmas and drug delivery systems. Then, the mechanism of microenvironment characteristics of bacterial infected lesions was elucidated to provide a strong theoretical basis for bacteria-targeting nanosystems design. In particular, the discussion focuses on the design principles of single-stimulus and dual-stimulus responsive nanosystems, as well as the use of endogenous stimulus-responsive nanosystems to deliver antimicrobial agents to target locations for combating bacterial infectious diseases. Finally, the challenges and prospects of endogenous stimulus-responsive nanosystems were summarized.
Collapse
Affiliation(s)
- Niuniu Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Institute of Nutrition and Health,Huazhong Agricultural University, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Mengyuan Sun
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Huixin Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Danlei Hu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Aoxue Zhang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Suliman Khan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Zhen Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Institute of Nutrition and Health,Huazhong Agricultural University, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
24
|
Liu H, Xie T, Wang J, Wang X, Han J, Huang Z, Jiang L, Nie Z. In situ analysis of metabolic changes under hypoxic-ischemic encephalopathy via MALDI mass spectrometry imaging. Talanta 2024; 268:125306. [PMID: 37839325 DOI: 10.1016/j.talanta.2023.125306] [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: 06/01/2023] [Revised: 09/28/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a leading cause of neurological disability and even more serious fetal or neonatal asphyxia death. As the therapeutic time window is limited and timely intervention could have a better prognosis, elucidating the mechanisms underlying HIE and developing novel therapeutic strategies is of great importance. In the present study, 1, 5-Diaminonaphthalene hydrochloride (1, 5-DANHCl) assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) was applied to the neonatal rat model of HIE to investigate metabolic changes during hypoxic-ischemic period. Seventy-three metabolites involved in various metabolic pathways such as glycolysis, tricarboxylic acid (TCA) cycle, nucleoside metabolism, lipid metabolism, oxidative stress and ionic homeostasis demonstrated significant changes. It is worth mentioning that we have detected neutral triglycerides (TGs) that are difficult to ionize and observed their accumulation in the ischemic region, which has been rarely reported in previous studies. The results not only help us discover biomarkers but also provide new insights into its mechanism for us to understand the pathological and physiological processes of the disease.
Collapse
Affiliation(s)
- Huihui Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ting Xie
- Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China; Gannna Innovation and Translational Medicine Research Institute, Ganzhou, Jiangxi Province, 341000, China
| | - Jiyun Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Han
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhihua Huang
- Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000, China; Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China.
| | - Lixia Jiang
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China; Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000, China.
| | - Zongxiu Nie
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
25
|
Stanley S, Wang X, Liu Q, Kwon YY, Frey AM, Hicks ND, Vickers AJ, Hui S, Fortune SM. Ongoing evolution of the Mycobacterium tuberculosis lactate dehydrogenase reveals the pleiotropic effects of bacterial adaption to host pressure. PLoS Pathog 2024; 20:e1012050. [PMID: 38422159 DOI: 10.1371/journal.ppat.1012050] [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: 10/20/2023] [Revised: 03/12/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
The bacterial determinants that facilitate Mycobacterium tuberculosis (Mtb) adaptation to the human host environment are poorly characterized. We have sought to decipher the pressures facing the bacterium in vivo by assessing Mtb genes that are under positive selection in clinical isolates. One of the strongest targets of selection in the Mtb genome is lldD2, which encodes a quinone-dependent L-lactate dehydrogenase (LldD2) that catalyzes the oxidation of lactate to pyruvate. Lactate accumulation is a salient feature of the intracellular environment during infection and lldD2 is essential for Mtb growth in macrophages. We determined the extent of lldD2 variation across a set of global clinical isolates and defined how prevalent mutations modulate Mtb fitness. We show the stepwise nature of lldD2 evolution that occurs as a result of ongoing lldD2 selection in the background of ancestral lineage-defining mutations and demonstrate that the genetic evolution of lldD2 additively augments Mtb growth in lactate. Using quinone-dependent antibiotic susceptibility as a functional reporter, we also find that the evolved lldD2 mutations functionally increase the quinone-dependent activity of LldD2. Using 13C-lactate metabolic flux tracing, we find that lldD2 is necessary for robust incorporation of lactate into central carbon metabolism. In the absence of lldD2, label preferentially accumulates in dihydroxyacetone phosphate (DHAP) and glyceraldehyde-3-phosphate (G3P) and is associated with a discernible growth defect, providing experimental evidence for accrued lactate toxicity via the deleterious buildup of sugar phosphates. The evolved lldD2 variants increase lactate incorporation to pyruvate while altering triose phosphate flux, suggesting both an anaplerotic and detoxification benefit to lldD2 evolution. We further show that the mycobacterial cell is transcriptionally sensitive to the changes associated with altered lldD2 activity which affect the expression of genes involved in cell wall lipid metabolism and the ESX- 1 virulence system. Together, these data illustrate a multifunctional role of LldD2 that provides context for the selective advantage of lldD2 mutations in adapting to host stress.
Collapse
Affiliation(s)
- Sydney Stanley
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Xin Wang
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Qingyun Liu
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Young Yon Kwon
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Abigail M Frey
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Nathan D Hicks
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Andrew J Vickers
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
26
|
Xia A, Wan J, Li X, Quan J, Chen X, Xu Z, Jiao X. M. tb Rv0927c suppresses the activation of HIF-1α pathway through VHL-mediated ubiquitination and NF-κB/COX-2 pathway to enhance mycobacteria survival. Microbiol Res 2024; 278:127529. [PMID: 37922696 DOI: 10.1016/j.micres.2023.127529] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/24/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
Mycobacterium tuberculosis (M. tuberculosis), the causative agent of tuberculosis, employs various effector proteins to target and modulate host defenses. Our previous study showed that M. tuberculosis protein Rv0927c can promote the survival of intracellular mycobacteria, but the underlying mechanisms remain poorly understood. Here, we found that Rv0927c inhibited Mycobacterium smegmatis (M. smegmatis) induced hypoxia-inducible factor-1α (HIF-1α) activation in macrophages, and HIF-1α is required for Rv0927c to promote mycobacteria survival. Western blot analysis showed that Rv0927c promoted the proteasomal degradation of HIF-1α via Von Hippel-Lindau (VHL)-mediated ubiquitination and inhibited the nuclear localization of HIF-1α through the NF-κB/COX-2 pathway, thereby suppressing HIF-1α pathway activation. Furthermore, Rv0927c suppressed the host glycolytic metabolism, which is known to be regulated by HIF-1α and depended on the glycolysis process to promote mycobacterial survival. Our findings provide evidence that Rv0927c inhibits the activation of HIF-1α pathway, allowing pathogens to evade host immune responses, suggesting that targeting Rv0927c or HIF-1α might be a potential anti-tuberculosis therapy.
Collapse
Affiliation(s)
- Aihong Xia
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jiaxu Wan
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Xin Li
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Juanjuan Quan
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
27
|
Quadir N, Shariq M, Sheikh JA, Singh J, Sharma N, Hasnain SE, Ehtesham NZ. Mycobacterium tuberculosis protein MoxR1 enhances virulence by inhibiting host cell death pathways and disrupting cellular bioenergetics. Virulence 2023; 14:2180230. [PMID: 36799069 PMCID: PMC9980616 DOI: 10.1080/21505594.2023.2180230] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Mycobacterium tuberculosis (M. tb) utilizes the multifunctionality of its protein factors to deceive the host. The unabated global incidence and prevalence of tuberculosis (TB) and the emergence of multidrug-resistant strains warrant the discovery of novel drug targets that can be exploited to manage TB. This study reports the role of M. tb AAA+ family protein MoxR1 in regulating host-pathogen interaction and immune system functions. We report that MoxR1 binds to TLR4 in macrophage cells and further reveal how this signal the release of proinflammatory cytokines. We show that MoxR1 activates the PI3K-AKT-MTOR signalling cascade by inhibiting the autophagy-regulating kinase ULK1 by potentiating its phosphorylation at serine 757, leading to its suppression. Using autophagy-activating and repressing agents such as rapamycin and bafilomycin A1 suggested that MoxR1 inhibits autophagy flux by inhibiting autophagy initiation. MoxR1 also inhibits apoptosis by suppressing the expression of MAPK JNK1/2 and cFOS, which play critical roles in apoptosis induction. Intriguingly, MoxR1 also induced robust disruption of cellular bioenergetics by metabolic reprogramming to rewire the citric acid cycle intermediates, as evidenced by the lower levels of citric acid and electron transport chain enzymes (ETC) to dampen host defence. These results point to a multifunctional role of M. tb MoxR1 in dampening host defences by inhibiting autophagy, apoptosis, and inducing metabolic reprogramming. These mechanistic insights can be utilized to devise strategies to combat TB and better understand survival tactics by intracellular pathogens.
Collapse
Affiliation(s)
- Neha Quadir
- National Institute of Pathology, ICMR, Safdarjung Hospital Campus, New Delhi, India,Institute of Molecular Medicine, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| | - Mohd. Shariq
- National Institute of Pathology, ICMR, Safdarjung Hospital Campus, New Delhi, India
| | | | - Jasdeep Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| | - Neha Sharma
- National Institute of Pathology, ICMR, Safdarjung Hospital Campus, New Delhi, India
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India,Department of Life Science,School of Basic Science and Research, Sharda University, Greater Noida, India,CONTACT Seyed Ehtesham Hasnain
| | - Nasreen Zafar Ehtesham
- National Institute of Pathology, ICMR, Safdarjung Hospital Campus, New Delhi, India,Nazreen Zafar Ehtesham
| |
Collapse
|
28
|
Rai MK, Yadav S, Jain A, Singh K, Kumar A, Raj R, Dubey D, Singh H, Guleria A, Chaturvedi S, Khan AR, Nath A, Misra DP, Agarwal V, Kumar D. Clinical metabolomics by NMR revealed serum metabolic signatures for differentiating sarcoidosis from tuberculosis. Metabolomics 2023; 19:92. [PMID: 37940751 DOI: 10.1007/s11306-023-02052-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/20/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Pulmonary sarcoidosis (SAR) and tuberculosis (TB) are two granulomatous lung-diseases and often pose a diagnostic challenge to a treating physicians. OBJECTIVE The present study aims to explore the diagnostic potential of NMR based serum metabolomics approach to differentiate SAR from TB. MATERIALS AND METHOD The blood samples were obtained from three study groups: SAR (N = 35), TB (N = 28) and healthy normal subjects (NC, N = 56) and their serum metabolic profiles were measured using 1D 1H CPMG (Carr-Purcell-Meiboom-Gill) NMR spectra recorded at 800 MHz NMR spectrometer. The quantitative metabolic profiles were compared employing a combination of univariate and multivariate statistical analysis methods and evaluated for their diagnostic potential using receiver operating characteristic (ROC) curve analysis. RESULTS Compared to SAR, the sera of TB patients were characterized by (a) elevated levels of lactate, acetate, 3-hydroxybutyrate (3HB), glutamate and succinate (b) decreased levels of glucose, citrate, pyruvate, glutamine, and several lipid and membrane metabolites (such as very-low/low density lipoproteins (VLDL/LDL), polyunsaturated fatty acids, etc.). CONCLUSION The metabolic disturbances not only found to be well in concordance with various previous reports, these further demonstrated very high sensitivity and specificity to distinguish SAR from TB patients suggesting serum metabolomics analysis can serve as surrogate method in the diagnosis and clinical management of SAR.
Collapse
Affiliation(s)
- Mohit Kumar Rai
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India
| | - Sachin Yadav
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India
- Department of Chemistry, Integral University, Lucknow, UP, 226026, India
| | - Avinash Jain
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India.
- Department of Clinical Immunology and Rheumatology, SMS Medical College, Jaipur, India.
| | - Kritika Singh
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India
| | - Amit Kumar
- Centre of Biomedical Research (CBMR), Lucknow, UP, 226014, India
| | - Ritu Raj
- Centre of Biomedical Research (CBMR), Lucknow, UP, 226014, India
| | - Durgesh Dubey
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India
- Centre of Biomedical Research (CBMR), Lucknow, UP, 226014, India
| | - Harshit Singh
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India
- Immuno Biology Lab, Translational Health Science and Technology Institute, Faridabad, HR, 121001, India
| | - Anupam Guleria
- Centre of Biomedical Research (CBMR), Lucknow, UP, 226014, India
| | - Saurabh Chaturvedi
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Delhi Pharmaceutical Sciences and Research University, Sector III, Pushp Vihar, M.B. Road, New Delhi, 110017, India
| | - Abdul Rahman Khan
- Department of Chemistry, Integral University, Lucknow, UP, 226026, India
| | - Alok Nath
- Department of Pulmonary Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India
| | - Durga Prasanna Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India
| | - Vikas Agarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, UP, 226014, India.
| | - Dinesh Kumar
- Centre of Biomedical Research (CBMR), Lucknow, UP, 226014, India.
| |
Collapse
|
29
|
Sinton MC, Chandrasegaran PRG, Capewell P, Cooper A, Girard A, Ogunsola J, Perona-Wright G, M Ngoyi D, Kuispond N, Bucheton B, Camara M, Kajimura S, Bénézech C, Mabbott NA, MacLeod A, Quintana JF. IL-17 signalling is critical for controlling subcutaneous adipose tissue dynamics and parasite burden during chronic murine Trypanosoma brucei infection. Nat Commun 2023; 14:7070. [PMID: 37923768 PMCID: PMC10624677 DOI: 10.1038/s41467-023-42918-8] [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: 05/26/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023] Open
Abstract
In the skin, Trypanosoma brucei colonises the subcutaneous white adipose tissue, and is proposed to be competent for forward transmission. The interaction between parasites, adipose tissue, and the local immune system is likely to drive the adipose tissue wasting and weight loss observed in cattle and humans infected with T. brucei. However, mechanistically, events leading to subcutaneous white adipose tissue wasting are not fully understood. Here, using several complementary approaches, including mass cytometry by time of flight, bulk and single cell transcriptomics, and in vivo genetic models, we show that T. brucei infection drives local expansion of several IL-17A-producing cells in the murine WAT, including TH17 and Vγ6+ cells. We also show that global IL-17 deficiency, or deletion of the adipocyte IL-17 receptor protect from infection-induced WAT wasting and weight loss. Unexpectedly, we find that abrogation of adipocyte IL-17 signalling results in a significant accumulation of Dpp4+ Pi16+ interstitial preadipocytes and increased extravascular parasites in the WAT, highlighting a critical role for IL-17 signalling in controlling preadipocyte fate, subcutaneous WAT dynamics, and local parasite burden. Taken together, our study highlights the central role of adipocyte IL-17 signalling in controlling WAT responses to infection, suggesting that adipocytes are critical coordinators of tissue dynamics and immune responses to T. brucei infection.
Collapse
Affiliation(s)
- Matthew C Sinton
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK.
- Division of Cardiovascular Science, University of Manchester, Manchester, UK.
| | - Praveena R G Chandrasegaran
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Paul Capewell
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Anneli Cooper
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Alex Girard
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - John Ogunsola
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Georgia Perona-Wright
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Dieudonné M Ngoyi
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of Congo
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Nono Kuispond
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of Congo
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Bruno Bucheton
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
- Institut de Recherche pour le Développement, Unité Mixte de Recherche IRD-CIRAD 177, Campus International de Baillarguet, Montpellier, France
| | - Mamadou Camara
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine, Ministère de la Santé, Conakry, Guinea
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Cécile Bénézech
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Juan F Quintana
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK.
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
- Division of Immunology, Immunity to Infection and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
30
|
Beam JE, Wagner NJ, Lu KY, Parsons JB, Fowler VG, Rowe SE, Conlon BP. Inflammasome-mediated glucose limitation induces antibiotic tolerance in Staphylococcus aureus. iScience 2023; 26:107942. [PMID: 37790275 PMCID: PMC10543182 DOI: 10.1016/j.isci.2023.107942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/27/2023] [Accepted: 09/13/2023] [Indexed: 10/05/2023] Open
Abstract
Staphylococcus aureus is a leading human pathogen that frequently causes relapsing infections. The failure of antibiotics to eradicate infection contributes to infection relapse. Host-pathogen interactions have a substantial impact on antibiotic susceptibility and the formation of antibiotic tolerant cells. In this study, we interrogate how a major S. aureus virulence factor, α-toxin, interacts with macrophages to alter the microenvironment of the pathogen, thereby influencing its susceptibility to antibiotics. We find α-toxin-mediated activation of the NLRP3 inflammasome induces antibiotic tolerance. Induction of tolerance is driven by increased glycolysis in the host cells, resulting in glucose limitation and ATP depletion in S. aureus. Additionally, inhibition of NLRP3 activation improves antibiotic efficacy in vitro and in vivo, suggesting that this strategy has potential as a host-directed therapeutic to improve outcomes. Our findings identify interactions between S. aureus and the host that result in metabolic crosstalk that can determine the outcome of antimicrobial therapy.
Collapse
Affiliation(s)
- Jenna E. Beam
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nikki J. Wagner
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kuan-Yi Lu
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joshua B. Parsons
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Vance G. Fowler
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian P. Conlon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
31
|
Stanley S, Wang X, Liu Q, Kwon YY, Frey AM, Hicks ND, Vickers AJ, Hui S, Fortune SM. Ongoing evolution of the Mycobacterium tuberculosis lactate dehydrogenase reveals the pleiotropic effects of bacterial adaption to host pressure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561592. [PMID: 37873410 PMCID: PMC10592758 DOI: 10.1101/2023.10.09.561592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The bacterial determinants that facilitate Mycobacterium tuberculosis (Mtb) adaptation to the human host environment are poorly characterized. We have sought to decipher the pressures facing the bacterium in vivo by assessing Mtb genes that are under positive selection in clinical isolates. One of the strongest targets of selection in the Mtb genome is lldD2 , which encodes a quinone-dependent L-lactate dehydrogenase (LldD2) that catalyzes the oxidation of lactate to pyruvate. Lactate accumulation is a salient feature of the intracellular environment during infection and lldD2 is essential for Mtb growth in macrophages. We determined the extent of lldD2 variation across a set of global clinical isolates and defined how prevalent mutations modulates Mtb fitness. We show the stepwise nature of lldD2 evolution that occurs as a result of ongoing lldD2 selection in the background of ancestral lineage defining mutations and demonstrate that the genetic evolution of lldD2 additively augments Mtb growth in lactate. Using quinone-dependent antibiotic susceptibility as a functional reporter, we also find that the evolved lldD2 mutations functionally increase the quinone-dependent activity of LldD2. Using 13 C-lactate metabolic flux tracing, we find that lldD2 is necessary for robust incorporation of lactate into central carbon metabolism. In the absence of lldD2 , label preferentially accumulates in methylglyoxal precursors dihydroxyacetone phosphate (DHAP) and glyceraldehyde-3-phosphate (G3P) and is associated with a discernible growth defect, providing experimental evidence for accumulated lactate toxicity via a methylglyoxal pathway that has been proposed previously. The evolved lldD2 variants increase lactate incorporation to pyruvate but also alter flux in the methylglyoxal pathway, suggesting both an anaplerotic and detoxification benefit to lldD2 evolution. We further show that the mycobacterial cell is transcriptionally sensitive to the changes associated with altered lldD2 activity which affect the expression of genes involved in cell wall lipid metabolism and the ESX-1 virulence system. Together, these data illustrate a multifunctional role of LldD2 that provide context for the selective advantage of lldD2 mutations in adapting to host stress.
Collapse
|
32
|
Ellzey LM, Patrick KL, Watson RO. Mitochondrial reactive oxygen species: double agents in Mycobacterium tuberculosis infection. Curr Opin Immunol 2023; 84:102366. [PMID: 37453340 DOI: 10.1016/j.coi.2023.102366] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/23/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
In addition to housing the major energy-producing pathways in cells, mitochondria are active players in innate immune responses. One critical way mitochondria fulfill this role is by releasing damage-associated molecular patterns (mtDAMPs) that are recognized by innate sensors to activate pathways including, but not limited to, cytokine expression, selective autophagy, and cell death. Mitochondrial reactive oxygen species (mtROS) is a multifunctional mtDAMP linked to pro- and antimicrobial immune outcomes. Formed as a by-product of energy generation, mtROS links mitochondrial metabolism with downstream innate immune responses. As a result, altered cellular metabolism can change mtROS levels and impact downstream antimicrobial responses in a variety of ways. MtROS has emerged as a particularly important mediator of pathogenesis during infection with Mycobacterium tuberculosis (Mtb), an intracellular bacterial pathogen that continues to pose a significant threat to global public health. Here, we will summarize how Mtb modulates mtROS levels in infected macrophages and how mtROS dictates Mtb infection outcomes by controlling inflammation, lipid peroxidation, and cell death. We propose that mtROS may serve as a biomarker to predict tuberculosis patient outcomes and/or a target for host-directed therapeutics.
Collapse
Affiliation(s)
- Lily M Ellzey
- Interdiscplinary Graduate Program in Genetics and Genomics, Texas A&M University, United States; Department of Microbial Pathogenesis and Immunology, Texas A&M University School of Medicine, United States
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M University School of Medicine, United States
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M University School of Medicine, United States.
| |
Collapse
|
33
|
Pacl HT, Chinta KC, Reddy VP, Nadeem S, Sevalkar RR, Nargan K, Lumamba K, Naidoo T, Glasgow JN, Agarwal A, Steyn AJC. NAD(H) homeostasis underlies host protection mediated by glycolytic myeloid cells in tuberculosis. Nat Commun 2023; 14:5472. [PMID: 37673914 PMCID: PMC10482943 DOI: 10.1038/s41467-023-40545-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: 10/14/2022] [Accepted: 07/31/2023] [Indexed: 09/08/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) disrupts glycolytic flux in infected myeloid cells through an unclear mechanism. Flux through the glycolytic pathway in myeloid cells is inextricably linked to the availability of NAD+, which is maintained by NAD+ salvage and lactate metabolism. Using lung tissue from tuberculosis (TB) patients and myeloid deficient LDHA (LdhaLysM-/-) mice, we demonstrate that glycolysis in myeloid cells is essential for protective immunity in TB. Glycolytic myeloid cells are essential for the early recruitment of multiple classes of immune cells and IFNγ-mediated protection. We identify NAD+ depletion as central to the glycolytic inhibition caused by Mtb. Lastly, we show that the NAD+ precursor nicotinamide exerts a host-dependent, antimycobacterial effect, and that nicotinamide prophylaxis and treatment reduce Mtb lung burden in mice. These findings provide insight into how Mtb alters host metabolism through perturbation of NAD(H) homeostasis and reprogramming of glycolysis, highlighting this pathway as a potential therapeutic target.
Collapse
Affiliation(s)
- Hayden T Pacl
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Krishna C Chinta
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vineel P Reddy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sajid Nadeem
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ritesh R Sevalkar
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kievershen Nargan
- Africa Health Research Institute, University of KwaZulu Natal, Durban, South Africa
| | - Kapongo Lumamba
- Africa Health Research Institute, University of KwaZulu Natal, Durban, South Africa
| | - Threnesan Naidoo
- Africa Health Research Institute, University of KwaZulu Natal, Durban, South Africa
- Department of Laboratory Medicine and Pathology, Walter Sisulu University, Eastern Cape, South Africa
| | - Joel N Glasgow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Department of Medicine, Division of Nephrology, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Adrie J C Steyn
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Africa Health Research Institute, University of KwaZulu Natal, Durban, South Africa.
- Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
34
|
Jaiswal S, Kumar S, Velarde de la Cruz E. Exploring the role of the protein tyrosine kinase a (PtkA) in mycobacterial intracellular survival. Tuberculosis (Edinb) 2023; 142:102398. [PMID: 37657276 DOI: 10.1016/j.tube.2023.102398] [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: 05/20/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023]
Abstract
Mycobacterium tuberculosis (Mtb) continues to define new paradigms of host-pathogen interaction. There are several host proteins known which are regulated by Mtb infection. The proteins which regulate host biological processes like apoptosis, cell processes, stress proteins, metabolic enzymes, etc. are targeted by the pathogens. Mtb proteins interact directly or indirectly with host proteins and play an important role in their persistence and intracellular growth. Mtb is an intracellular pathogen. It remains dormant for years within the host without activating its immune system. Mtb Protein tyrosine kinase (PtkA) regulates host anti-apoptotic protein, metabolic enzymes, and several other proteins that are involved in stress regulation, cell proliferation, protein folding, DNA repair, etc. PtkA regulates other mycobacterial proteins and plays an important role in its growth and survival. Here we summarized the current knowledge of PtkA and reviewed its role in mycobacterial intracellular survival as it regulates several other mycobacterial proteins and host proteins. PtkA regulates PtpA secretion which is essential for mycobacterial virulence and could be used as an attractive drug target.
Collapse
Affiliation(s)
- Swati Jaiswal
- University of Massachusetts Chan Medical School, Worcester, United States.
| | | | | |
Collapse
|
35
|
Quan H, Chung H, Je S, Hong JJ, Kim BJ, Na YR, Seok SH. Pyruvate dehydrogenase kinase inhibitor dichloroacetate augments autophagy mediated constraining the replication of Mycobacteroides massiliense in macrophages. Microbes Infect 2023; 25:105139. [PMID: 37085043 DOI: 10.1016/j.micinf.2023.105139] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/25/2023] [Accepted: 04/11/2023] [Indexed: 04/23/2023]
Abstract
Increasing evidence indicates a strong interaction between cellular metabolism and innate macrophage immunity. Here, we show that the intracellular replication of Mycobacteroides massiliense in macrophages depends on host pyruvate dehydrogenase kinase (PDK) activity. Infection with M. massiliense induced a metabolic switch in macrophages by increasing glycolysis and decreasing oxidative phosphorylation. Treatment with dichloroacetate (DCA), a PDK inhibitor, converts this switch in M. massiliense-infected macrophages and restricts intracellular bacterial replication. Mechanistically, DCA resulted in AMPKα1 activation via increased AMP/ATP ratio, consequently inducing autophagy to constrain bacterial proliferation in the phagolysosome. This study suggests that the pharmacological inhibition of PDK could be a strategy for host-directed therapy to control virulent M. massiliense infections.
Collapse
Affiliation(s)
- Hailian Quan
- Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyewon Chung
- Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Republic of Korea; Bio-MAX Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sungmo Je
- Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, South Korea; KRIBB School of Bioscience, Korea University of Science & Technology (UST), Daejeon, South Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yi Rang Na
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Seung Hyeok Seok
- Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Yi J, Xiang J, Tang J. Exploring the microbiome: Uncovering the link with lung cancer and implications for diagnosis and treatment. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:161-170. [PMID: 39171127 PMCID: PMC11332872 DOI: 10.1016/j.pccm.2023.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Indexed: 08/23/2024]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Tobacco smoking and air pollution are believed to be responsible for more than 90% of lung cancers. Respiratory pathogens are also known to be associated with the initiation and development of lung cancer. Despite the fact that the bacterial biomass in the lungs is lower than that in the intestinal tract, emerging evidence indicates that the lung is colonized by a diverse array of microbes. However, there is limited knowledge regarding the role of dysbiosis of the lung microbiota in the progression of lung cancer. In this review, we summarize the current information about the relationship between the microbiome and lung cancer. The objective is to provide an overview of the core composition of the microbiota in lung cancer as well as the role of specific dysbiosis of the lung microbiota in the progression of lung cancer and treatment of the disease.
Collapse
Affiliation(s)
- Junqi Yi
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Juanjuan Xiang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410028, China
| | - Jingqun Tang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
37
|
Limón G, Samhadaneh NM, Pironti A, Darwin KH. Aldehyde accumulation in Mycobacterium tuberculosis with defective proteasomal degradation results in copper sensitivity. mBio 2023; 14:e0036323. [PMID: 37350636 PMCID: PMC10470581 DOI: 10.1128/mbio.00363-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/17/2023] [Indexed: 06/24/2023] Open
Abstract
Mycobacterium tuberculosis is a major human pathogen and the causative agent of tuberculosis disease. M. tuberculosis is able to persist in the face of host-derived antimicrobial molecules nitric oxide (NO) and copper (Cu). However, M. tuberculosis with defective proteasome activity is highly sensitive to NO and Cu, making the proteasome an attractive target for drug development. Previous work linked NO susceptibility with the accumulation of para-hydroxybenzaldehyde (pHBA) in M. tuberculosis mutants with defective proteasomal degradation. In this study, we found that pHBA accumulation was also responsible for Cu sensitivity in these strains. We showed that exogenous addition of pHBA to wild-type M. tuberculosis cultures sensitized bacteria to Cu to a degree similar to that of a proteasomal degradation mutant. We determined that pHBA reduced the production and function of critical Cu resistance proteins of the regulated in copper repressor (RicR) regulon. Furthermore, we extended these Cu-sensitizing effects to an aldehyde that M. tuberculosis may face within the macrophage. Collectively, this study is the first to mechanistically propose how aldehydes can render M. tuberculosis susceptible to an existing host defense and could support a broader role for aldehydes in controlling M. tuberculosis infections. IMPORTANCE M. tuberculosis is a leading cause of death by a single infectious agent, causing 1.5 million deaths annually. An effective vaccine for M. tuberculosis infections is currently lacking, and prior infection does not typically provide robust immunity to subsequent infections. Nonetheless, immunocompetent humans can control M. tuberculosis infections for decades. For these reasons, a clear understanding of how mammalian immunity inhibits mycobacterial growth is warranted. In this study, we show aldehydes can increase M. tuberculosis susceptibility to copper, an established antibacterial metal used by immune cells to control M. tuberculosis and other microbes. Given that activated macrophages produce increased amounts of aldehydes during infection, we propose host-derived aldehydes may help control bacterial infections, making aldehydes a previously unappreciated antimicrobial defense.
Collapse
Affiliation(s)
- Gina Limón
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Nora M. Samhadaneh
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Microbial Computational Genomic Core Lab, New York University Grossman School of Medicine, New York, New York, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Microbial Computational Genomic Core Lab, New York University Grossman School of Medicine, New York, New York, USA
| | - K. Heran Darwin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
38
|
Wang Y, He X, Zheng D, He Q, Sun L, Jin J. Integration of Metabolomics and Transcriptomics Reveals Major Metabolic Pathways and Potential Biomarkers Involved in Pulmonary Tuberculosis and Pulmonary Tuberculosis-Complicated Diabetes. Microbiol Spectr 2023; 11:e0057723. [PMID: 37522815 PMCID: PMC10434036 DOI: 10.1128/spectrum.00577-23] [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: 02/10/2023] [Accepted: 06/07/2023] [Indexed: 08/01/2023] Open
Abstract
Pulmonary tuberculosis (PTB) and diabetes mellitus (DM) are common chronic diseases that threaten human health. Patients with DM are susceptible to PTB, an important factor that aggravates the complications of diabetes. However, the molecular regulatory mechanism underlying the susceptibility of patients with DM to PTB infection remains unknown. In this study, healthy subjects, patients with primary PTB, and patients with primary PTB complicated by DM were recruited according to inclusion and exclusion criteria. Peripheral whole blood was collected, and alteration profiles and potential molecular mechanisms were further analyzed using integrated bioinformatics analysis of metabolomics and transcriptomics. Transcriptional data revealed that lipocalin 2 (LCN2), defensin alpha 1 (DEFA1), peptidoglycan recognition protein 1 (PGLYRP1), and integrin subunit alpha 2b (ITGA2B) were significantly upregulated, while chloride intracellular channel 3 (CLIC3) was significantly downregulated in the group with PTB and DM (PTB_DM) in contrast to the healthy control (HC) group. Additionally, the interleukin 17 (IL-17), phosphatidylinositol 3-kinase (PI3K)-AKT, and peroxisome proliferator-activated receptor (PPAR) signaling pathways are important for PTB infection and regulation of PTB-complicated diabetes. Metabolomic data showed that glycerophospholipid metabolism, carbon metabolism, and fat digestion and absorption processes were enriched in the differential metabolic analysis. Finally, integrated analysis of both metabolomic and transcriptomic data indicated that the NOTCH1/JAK/STAT signaling pathway is important in PTB complicated by DM. In conclusion, PTB infection altered the transcriptional and metabolic profiles of patients with DM. Metabolomic and transcriptomic changes were highly correlated in PTB patients with DM. Peripheral metabolite levels may be used as biomarkers for PTB management in patients with DM. IMPORTANCE The comorbidity of diabetes mellitus (DM) significantly increases the risk of tuberculosis infection and adverse tuberculosis treatment outcomes. Most previous studies have focused on the relationship between the effect of blood glucose control and the outcome of antituberculosis treatment in pulmonary tuberculosis (PTB) with DM (PTB_DM); however, early prediction and the underlying molecular mechanism of susceptibility to PTB infection in patients with DM remain unclear. In this study, transcriptome sequencing and untargeted metabolomics were performed to elucidate the key molecules and signaling pathways involved in PTB infection and the susceptibility of patients with diabetes to PTB. Our findings contribute to the development of vital diagnostic biomarkers for PTB or PTB_DM and provide a comprehensive understanding of molecular regulation during disease progression.
Collapse
Affiliation(s)
- Yunguang Wang
- Department of Critical Care Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Xinxin He
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Danna Zheng
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, People’s Republic of China
| | - Qiang He
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, People’s Republic of China
| | - Lifang Sun
- Department of Tuberculosis, Affiliated Hangzhou Chest Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Department of Tuberculosis, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, People’s Republic of China
| | - Juan Jin
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
39
|
Maurice NM, Sadikot RT. Mitochondrial Dysfunction in Bacterial Infections. Pathogens 2023; 12:1005. [PMID: 37623965 PMCID: PMC10458073 DOI: 10.3390/pathogens12081005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Mitochondria are critical in numerous cellular processes, including energy generation. Bacterial pathogens target host cell mitochondria through various mechanisms to disturb the host response and improve bacterial survival. We review recent advances in the understanding of how bacteria cause mitochondrial dysfunction through perturbations in mitochondrial cell-death pathways, energy production, mitochondrial dynamics, mitochondrial quality control, DNA repair, and the mitochondrial unfolded protein response. We also briefly highlight possible therapeutic approaches aimed at restoring the host mitochondrial function as a novel strategy to enhance the host response to bacterial infection.
Collapse
Affiliation(s)
- Nicholas M. Maurice
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Ruxana T. Sadikot
- VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
40
|
Whittington AM, Turner FS, Baark F, Templeman S, Kirwan DE, Roufosse C, Krishnan N, Robertson BD, Chong DLW, Porter JC, Gilman RH, Friedland JS. An acidic microenvironment in Tuberculosis increases extracellular matrix degradation by regulating macrophage inflammatory responses. PLoS Pathog 2023; 19:e1011495. [PMID: 37418488 PMCID: PMC10355421 DOI: 10.1371/journal.ppat.1011495] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 07/19/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb) infection causes marked tissue inflammation leading to lung destruction and morbidity. The inflammatory extracellular microenvironment is acidic, however the effect of this acidosis on the immune response to M.tb is unknown. Using RNA-seq we show that acidosis produces system level transcriptional change in M.tb infected human macrophages regulating almost 4000 genes. Acidosis specifically upregulated extracellular matrix (ECM) degradation pathways with increased expression of Matrix metalloproteinases (MMPs) which mediate lung destruction in Tuberculosis. Macrophage MMP-1 and -3 secretion was increased by acidosis in a cellular model. Acidosis markedly suppresses several cytokines central to control of M.tb infection including TNF-α and IFN-γ. Murine studies demonstrated expression of known acidosis signaling G-protein coupled receptors OGR-1 and TDAG-8 in Tuberculosis which are shown to mediate the immune effects of decreased pH. Receptors were then demonstrated to be expressed in patients with TB lymphadenitis. Collectively, our findings show that an acidic microenvironment modulates immune function to reduce protective inflammatory responses and increase extracellular matrix degradation in Tuberculosis. Acidosis receptors are therefore potential targets for host directed therapy in patients.
Collapse
Affiliation(s)
| | - Frances S. Turner
- Edinburgh Genomics, University of Edinburgh, Edinburgh, United Kingdom
| | - Friedrich Baark
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Sam Templeman
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Daniela E. Kirwan
- Institute of Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - Candice Roufosse
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Nitya Krishnan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Brian D. Robertson
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Deborah L. W. Chong
- Institute of Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - Joanna C. Porter
- Centre for Inflammation & Tissue Repair, Respiratory Medicine, University College London, London, United Kingdom
| | - Robert H. Gilman
- Department of International Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jon S. Friedland
- Institute of Infection and Immunity, St. George’s, University of London, London, United Kingdom
| |
Collapse
|
41
|
Bisht MK, Dahiya P, Ghosh S, Mukhopadhyay S. The cause-effect relation of tuberculosis on incidence of diabetes mellitus. Front Cell Infect Microbiol 2023; 13:1134036. [PMID: 37434784 PMCID: PMC10330781 DOI: 10.3389/fcimb.2023.1134036] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/25/2023] [Indexed: 07/13/2023] Open
Abstract
Tuberculosis (TB) is one of the oldest human diseases and is one of the major causes of mortality and morbidity across the Globe. Mycobacterium tuberculosis (Mtb), the causal agent of TB is one of the most successful pathogens known to mankind. Malnutrition, smoking, co-infection with other pathogens like human immunodeficiency virus (HIV), or conditions like diabetes further aggravate the tuberculosis pathogenesis. The association between type 2 diabetes mellitus (DM) and tuberculosis is well known and the immune-metabolic changes during diabetes are known to cause increased susceptibility to tuberculosis. Many epidemiological studies suggest the occurrence of hyperglycemia during active TB leading to impaired glucose tolerance and insulin resistance. However, the mechanisms underlying these effects is not well understood. In this review, we have described possible causal factors like inflammation, host metabolic changes triggered by tuberculosis that could contribute to the development of insulin resistance and type 2 diabetes. We have also discussed therapeutic management of type 2 diabetes during TB, which may help in designing future strategies to cope with TB-DM cases.
Collapse
Affiliation(s)
- Manoj Kumar Bisht
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Priyanka Dahiya
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Sudip Ghosh
- Molecular Biology Unit, Indian Council of Medical Research (ICMR)-National Institute of Nutrition, Jamai Osmania PO, Hyderabad, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| |
Collapse
|
42
|
Nguyen MT, Hu Z, Mohammad M, Schöttler H, Niemann S, Schultz M, Barczyk-Kahlert K, Jin T, Hayen H, Herrmann M. Bacterial Lipoproteins Shift Cellular Metabolism to Glycolysis in Macrophages Causing Bone Erosion. Microbiol Spectr 2023; 11:e0429322. [PMID: 37191536 PMCID: PMC10269925 DOI: 10.1128/spectrum.04293-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/25/2023] [Indexed: 05/17/2023] Open
Abstract
Belonging to a group of membrane proteins, bacterial lipoproteins (LPPs) are defined by a unique lipid structure at their N-terminus providing the anchor in the bacterial cell membrane. In Gram-positive bacteria, LPPs play a key role in host immune activation triggered through a Toll-like receptor 2 (TLR2)-mediated action resulting in macrophage stimulation and subsequent tissue damage demonstrated in in vivo experimental models. Yet the physiologic links between LPP activation, cytokine release, and any underlying switches in cellular metabolism remain unclear. In this study, we demonstrate that Staphylococcus aureus Lpl1 not only triggers cytokine production but also confers a shift toward fermentative metabolism in bone marrow-derived macrophages (BMDMs). Lpl1 consists of di- and tri-acylated LPP variants; hence, the synthetic P2C and P3C, mimicking di-and tri-acylated LPPs, were employed to reveal their effect on BMDMs. Compared to P3C, P2C was found to shift the metabolism of BMDMs and the human mature monocytic MonoMac 6 (MM6) cells more profoundly toward the fermentative pathway, as indicated by lactate accumulation, glucose consumption, pH reduction, and oxygen consumption. In vivo, P2C caused more severe joint inflammation, bone erosion, and lactate and malate accumulation than P3C. These observed P2C effects were completely abrogated in monocyte/macrophage-depleted mice. Taken together, these findings now solidly confirm the hypothesized link between LPP exposure, a macrophage metabolic shift toward fermentation, and ensuing bone destruction. IMPORTANCE Osteomyelitis caused by S. aureus is a severe infection of the bone, typically associated with severe bone function impairment, therapeutic failure, high morbidity, invalidity, and occasionally even death. The hallmark of staphylococcal osteomyelitis is the destruction of the cortical bone structures, yet the mechanisms contributing to this pathology are hitherto poorly understood. One bacterial membrane constituent found in all bacteria is bacterial lipoproteins (LPPs). Previously, we have shown that injection of purified S. aureus LPPs into wild-type mouse knee joints caused a TLR2-dependent chronic destructive arthritis but failed to elicit such effect in monocyte/macrophage-depleted mice. This observation stirred our interest in investigating the interaction of LPPs and macrophages and analyzing the underlying physiological mechanisms. This ascertainment of LPP-induced changes in the physiology of macrophages provides an important clue in the understanding of the mechanisms of bone disintegration, opening novel avenues to manage the course of S. aureus disease.
Collapse
Affiliation(s)
- Minh-Thu Nguyen
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Zhicheng Hu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hannah Schöttler
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Michelle Schultz
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Heiko Hayen
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Mathias Herrmann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| |
Collapse
|
43
|
Salido S, Alejo-Armijo A, Altarejos J. Synthesis and hLDH Inhibitory Activity of Analogues to Natural Products with 2,8-Dioxabicyclo[3.3.1]nonane Scaffold. Int J Mol Sci 2023; 24:9925. [PMID: 37373073 DOI: 10.3390/ijms24129925] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Human lactate dehydrogenase (hLDH) is a tetrameric enzyme present in almost all tissues. Among its five different isoforms, hLDHA and hLDHB are the predominant ones. In the last few years, hLDHA has emerged as a therapeutic target for the treatment of several kinds of disorders, including cancer and primary hyperoxaluria. hLDHA inhibition has been clinically validated as a safe therapeutic method and clinical trials using biotechnological approaches are currently being evaluated. Despite the well-known advantages of pharmacological treatments based on small-molecule drugs, few compounds are currently in preclinical stage. We have recently reported the detection of some 2,8-dioxabicyclo[3.3.1]nonane core derivatives as new hLDHA inhibitors. Here, we extended our work synthesizing a large number of derivatives (42-70) by reaction between flavylium salts (27-35) and several nucleophiles (36-41). Nine 2,8-dioxabicyclo[3.3.1]nonane derivatives showed IC50 values lower than 10 µM against hLDHA and better activity than our previously reported compound 2. In order to know the selectivity of the synthesized compounds against hLDHA, their hLDHB inhibitory activities were also measured. In particular, compounds 58, 62a, 65b, and 68a have shown the lowest IC50 values against hLDHA (3.6-12.0 µM) and the highest selectivity rate (>25). Structure-activity relationships have been deduced. Kinetic studies using a Lineweaver-Burk double-reciprocal plot have indicated that both enantiomers of 68a and 68b behave as noncompetitive inhibitors on hLDHA enzyme.
Collapse
Affiliation(s)
- Sofía Salido
- Departamento de Química Inorgánica y Orgánica, Facultad de Ciencias Experimentales, Universidad de Jaén, Campus de Excelencia Internacional Agroalimentario ceiA3, 23071 Jaén, Spain
| | - Alfonso Alejo-Armijo
- Departamento de Química Inorgánica y Orgánica, Facultad de Ciencias Experimentales, Universidad de Jaén, Campus de Excelencia Internacional Agroalimentario ceiA3, 23071 Jaén, Spain
| | - Joaquín Altarejos
- Departamento de Química Inorgánica y Orgánica, Facultad de Ciencias Experimentales, Universidad de Jaén, Campus de Excelencia Internacional Agroalimentario ceiA3, 23071 Jaén, Spain
| |
Collapse
|
44
|
Polinário G, Primo LMDG, Rosa MABC, Dett FHM, Barbugli PA, Roque-Borda CA, Pavan FR. Antimicrobial peptides as drugs with double response against Mycobacterium tuberculosis coinfections in lung cancer. Front Microbiol 2023; 14:1183247. [PMID: 37342560 PMCID: PMC10277934 DOI: 10.3389/fmicb.2023.1183247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Tuberculosis and lung cancer are, in many cases, correlated diseases that can be confused because they have similar symptoms. Many meta-analyses have proven that there is a greater chance of developing lung cancer in patients who have active pulmonary tuberculosis. It is, therefore, important to monitor the patient for a long time after recovery and search for combined therapies that can treat both diseases, as well as face the great problem of drug resistance. Peptides are molecules derived from the breakdown of proteins, and the membranolytic class is already being studied. It has been proposed that these molecules destabilize cellular homeostasis, performing a dual antimicrobial and anticancer function and offering several possibilities of adaptation for adequate delivery and action. In this review, we focus on two important reason for the use of multifunctional peptides or peptides, namely the double activity and no harmful effects on humans. We review some of the main antimicrobial and anti-inflammatory bioactive peptides and highlight four that have anti-tuberculosis and anti-cancer activity, which may contribute to obtaining drugs with this dual functionality.
Collapse
Affiliation(s)
- Giulia Polinário
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | | | | | | | - Paula Aboud Barbugli
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | | | - Fernando Rogério Pavan
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
45
|
Weerasinghe H, Simm C, Djajawi TM, Tedja I, Lo TL, Simpson DS, Shasha D, Mizrahi N, Olivier FAB, Speir M, Lawlor KE, Ben-Ami R, Traven A. Candida auris uses metabolic strategies to escape and kill macrophages while avoiding robust activation of the NLRP3 inflammasome response. Cell Rep 2023; 42:112522. [PMID: 37204928 DOI: 10.1016/j.celrep.2023.112522] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/28/2023] [Accepted: 05/01/2023] [Indexed: 05/21/2023] Open
Abstract
Metabolic adaptations regulate the response of macrophages to infection. The contributions of metabolism to macrophage interactions with the emerging fungal pathogen Candida auris are poorly understood. Here, we show that C. auris-infected macrophages undergo immunometabolic reprogramming and increase glycolysis but fail to activate a strong interleukin (IL)-1β cytokine response or curb C. auris growth. Further analysis shows that C. auris relies on its own metabolic capacity to escape from macrophages and proliferate in vivo. Furthermore, C. auris kills macrophages by triggering host metabolic stress through glucose starvation. However, despite causing macrophage cell death, C. auris does not trigger robust activation of the NLRP3 inflammasome. Consequently, inflammasome-dependent responses remain low throughout infection. Collectively, our findings show that C. auris uses metabolic regulation to eliminate macrophages while remaining immunologically silent to ensure its own survival. Thus, our data suggest that host and pathogen metabolism could represent therapeutic targets for C. auris infections.
Collapse
Affiliation(s)
- Harshini Weerasinghe
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Claudia Simm
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Tirta Mario Djajawi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Irma Tedja
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Tricia L Lo
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Daniel S Simpson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - David Shasha
- Infectious Diseases Unit, Tel Aviv Sourasky Medical Centre, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naama Mizrahi
- Infectious Diseases Unit, Tel Aviv Sourasky Medical Centre, Tel Aviv, Israel
| | - Françios A B Olivier
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Mary Speir
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Ronen Ben-Ami
- Infectious Diseases Unit, Tel Aviv Sourasky Medical Centre, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ana Traven
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
46
|
Dhiman A, Talukdar S, Chaubey GK, Dilawari R, Modanwal R, Chaudhary S, Patidar A, Boradia VM, Kumbhar P, Raje CI, Raje M. Regulation of Macrophage Cell Surface GAPDH Alters LL-37 Internalization and Downstream Effects in the Cell. J Innate Immun 2023; 15:581-598. [PMID: 37080180 PMCID: PMC10315065 DOI: 10.1159/000530083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 03/03/2023] [Indexed: 04/22/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb), the major causative agent of tuberculosis, has evolved mechanisms to evade host defenses and persist within host cells. Host-directed therapies against infected cells are emerging as an effective option. Cationic host defense peptide LL-37 is known to internalize into cells and induce autophagy resulting in intracellular killing of M.tb. This peptide also regulates the immune system and interacts with the multifunctional protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH) inside macrophages. Our investigations revealed that GAPDH moonlights as a mononuclear cell surface receptor that internalizes LL-37. We confirmed that the surface levels of purinergic receptor 7, the receptor previously reported for this peptide, remained unaltered on M.tb infected macrophages. Upon infection or cellular activation with IFNγ, surface recruited GAPDH bound to and internalized LL-37 into endocytic compartments via a lipid raft-dependent process. We also discovered a role for GAPDH in LL-37-mediated autophagy induction and clearance of intracellular pathogens. In infected macrophages wherein GAPDH had been knocked down, we observed an inhibition of LL-37-mediated autophagy which was rescued by GAPDH overexpression. This process was dependent on intracellular calcium and p38 MAPK pathways. Our findings reveal a previously unknown process by which macrophages internalize an antimicrobial peptide via cell surface GAPDH and suggest a moonlighting role of GAPDH in regulating cellular phenotypic responses of LL-37 resulting in reduction of M.tb burden.
Collapse
Affiliation(s)
- Asmita Dhiman
- Institute of Microbial Technology, CSIR, Chandigarh, India
| | | | | | - Rahul Dilawari
- Institute of Microbial Technology, CSIR, Chandigarh, India
| | | | | | - Anil Patidar
- Institute of Microbial Technology, CSIR, Chandigarh, India
| | | | - Pradeep Kumbhar
- National Institute of Pharmaceutical Education and Research, Punjab, India
| | | | - Manoj Raje
- Institute of Microbial Technology, CSIR, Chandigarh, India
| |
Collapse
|
47
|
Yang B, Mukherjee T, Radhakrishnan R, Paidipally P, Ansari D, John S, Vankayalapati R, Tripathi D, Yi G. HIV-Differentiated Metabolite N-Acetyl-L-Alanine Dysregulates Human Natural Killer Cell Responses to Mycobacterium tuberculosis Infection. Int J Mol Sci 2023; 24:7267. [PMID: 37108430 PMCID: PMC10138430 DOI: 10.3390/ijms24087267] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) has latently infected over two billion people worldwide (LTBI) and caused ~1.6 million deaths in 2021. Human immunodeficiency virus (HIV) co-infection with Mtb will affect the Mtb progression and increase the risk of developing active tuberculosis by 10-20 times compared with HIV- LTBI+ patients. It is crucial to understand how HIV can dysregulate immune responses in LTBI+ individuals. Plasma samples collected from healthy and HIV-infected individuals were investigated using liquid chromatography-mass spectrometry (LC-MS), and the metabolic data were analyzed using the online platform Metabo-Analyst. ELISA, surface and intracellular staining, flow cytometry, and quantitative reverse-transcription PCR (qRT-PCR) were performed using standard procedures to determine the surface markers, cytokines, and other signaling molecule expressions. Seahorse extra-cellular flux assays were used to measure mitochondrial oxidative phosphorylation and glycolysis. Six metabolites were significantly less abundant, and two were significantly higher in abundance in HIV+ individuals compared with healthy donors. One of the HIV-upregulated metabolites, N-acetyl-L-alanine (ALA), inhibits pro-inflammatory cytokine IFN-γ production by the NK cells of LTBI+ individuals. ALA inhibits the glycolysis of LTBI+ individuals' NK cells in response to Mtb. Our findings demonstrate that HIV infection enhances plasma ALA levels to inhibit NK-cell-mediated immune responses to Mtb infection, offering a new understanding of the HIV-Mtb interaction and providing insights into the implication of nutrition intervention and therapy for HIV-Mtb co-infected patients.
Collapse
Affiliation(s)
- Baojun Yang
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Tanmoy Mukherjee
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Rajesh Radhakrishnan
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Padmaja Paidipally
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Danish Ansari
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Sahana John
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Ramakrishna Vankayalapati
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Deepak Tripathi
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Guohua Yi
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| |
Collapse
|
48
|
Zhang K, Sowers ML, Cherryhomes EI, Singh VK, Mishra A, Restrepo BI, Khan A, Jagannath C. Sirtuin-dependent metabolic and epigenetic regulation of macrophages during tuberculosis. Front Immunol 2023; 14:1121495. [PMID: 36993975 PMCID: PMC10040548 DOI: 10.3389/fimmu.2023.1121495] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/01/2023] [Indexed: 03/14/2023] Open
Abstract
Macrophages are the preeminent phagocytic cells which control multiple infections. Tuberculosis a leading cause of death in mankind and the causative organism Mycobacterium tuberculosis (MTB) infects and persists in macrophages. Macrophages use reactive oxygen and nitrogen species (ROS/RNS) and autophagy to kill and degrade microbes including MTB. Glucose metabolism regulates the macrophage-mediated antimicrobial mechanisms. Whereas glucose is essential for the growth of cells in immune cells, glucose metabolism and its downsteam metabolic pathways generate key mediators which are essential co-substrates for post-translational modifications of histone proteins, which in turn, epigenetically regulate gene expression. Herein, we describe the role of sirtuins which are NAD+-dependent histone histone/protein deacetylases during the epigenetic regulation of autophagy, the production of ROS/RNS, acetyl-CoA, NAD+, and S-adenosine methionine (SAM), and illustrate the cross-talk between immunometabolism and epigenetics on macrophage activation. We highlight sirtuins as emerging therapeutic targets for modifying immunometabolism to alter macrophage phenotype and antimicrobial function.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Mark L. Sowers
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ellie I. Cherryhomes
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Vipul K. Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Blanca I. Restrepo
- University of Texas Health Houston, School of Public Health, Brownsville, TX, United States
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| |
Collapse
|
49
|
Nissa MU, Pinto N, Ghosh B, Singh U, Goswami M, Srivastava S. Proteomic analysis of liver tissue reveals Aeromonas hydrophila infection mediated modulation of host metabolic pathways in Labeo rohita. J Proteomics 2023; 279:104870. [PMID: 36906258 DOI: 10.1016/j.jprot.2023.104870] [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: 12/29/2022] [Revised: 02/20/2023] [Accepted: 03/04/2023] [Indexed: 03/11/2023]
Abstract
Aeromonas hydrophila (Ah) is a Gram-negative bacterium and a serious global pathogen causing Motile Aeromonas Septicaemia (MAS) in fish leading to global loss in aquaculture. Investigation of the molecular alterations of host tissues such as liver could be a powerful approach to identify mechanistic and diagnostic immune signatures of disease pathogenesis. We performed a proteomic analysis of Labeo rohita liver tissue to examine the protein dynamics in the host cells during Ah infection. The proteomic data was acquired using two strategies; discovery and targeted proteomics. Label-free quantification was performed between Control and challenged group (AH) to identify the differentially expressed proteins (DEPs). A total of 2525 proteins were identified and 157 were DEPs. DEPs include metabolic enzymes (CS, SUCLG2), antioxidative proteins, cytoskeletal proteins and immune related proteins (TLR3, CLEC4E). Pathways like lysosome pathway, apoptosis, metabolism of xenobiotics by cytochrome P450 were enriched by downregulated proteins. However, upregulated proteins majorly mapped to innate immune system, signaling of B cell receptor, proteosome pathway, ribosome, carbon metabolism and protein processing in ER. Our study would help in exploring the role of Toll-like receptors, C-type lectins and, metabolic intermediates like citrate and succinate in Ah pathogenesis to understand the Ah infection in fish. SIGNIFICANCE: Bacterial diseases such as motile aeromonas septicaemia (MAS) are among the most serious problems in aquaculture industry. Small molecules that target the metabolism of the host have recently emerged as potential treatment possibilities in infectious diseases. However, the ability to develop new therapies is hampered due to lack of knowledge about pathogenesis mechanisms and host-pathogen interactions. We examined alterations in the host proteome during MAS caused by Aeromonas hydrophila (Ah) infection, in Labeo rohita liver tissue to find cellular proteins and processes affected by Ah infection. Upregulated proteins belong to innate immune system, signaling of B cell receptor, proteosome pathway, ribosome, carbon metabolism and protein processing. Our work is an important step towards leveraging host metabolism in targeting the disease by providing a bigger picture on proteome pathology correlation during Ah infection.
Collapse
Affiliation(s)
- Mehar Un Nissa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Nevil Pinto
- Central Institute of Fisheries Education, Indian Council of Agricultural Research, Versova, Mumbai, Maharashtra 400061, India
| | - Biplab Ghosh
- Regional Centre for Biotechnology, Faridabad 121001, India
| | - Urvi Singh
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, 110034, India
| | - Mukunda Goswami
- Central Institute of Fisheries Education, Indian Council of Agricultural Research, Versova, Mumbai, Maharashtra 400061, India.
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
50
|
Yang B, Mukherjee T, Radhakrishnan R, Paidipally P, Ansari D, John S, Vankayalapati R, Tripathi D, Yi G. HIV-differentiated metabolite N-Acetyl-L-Alanine dysregulates human natural killer cell responses to Mycobacterium tuberculosis infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530445. [PMID: 36909560 PMCID: PMC10002710 DOI: 10.1101/2023.02.28.530445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Background Mycobacterium tuberculosis ( Mtb ) has latently infected over two billion people worldwide (LTBI) and causes 1.8 million deaths each year. Human immunodeficiency virus (HIV) co-infection with Mtb will affect the Mtb progression and increase the risk of developing active tuberculosis by 10-20 times compared to the HIV-LTBI+ patients. It is crucial to understand how HIV can dysregulate immune responses in LTBI+ individuals. Methods Plasma samples collected from healthy and HIV-infected individuals were investigated by liquid chromatography-mass spectrometry (LC-MS), and the metabolic data were analyzed using an online platform Metabo-Analyst. ELISA, surface and intracellular staining, flow cytometry, quantitative reverse transcription PCR (qRT-PCR) were performed by standard procedure to determine the surface markers, cytokines and other signaling molecule expression. Seahorse extra cellular flux assays were used to measure the mitochondrial oxidative phosphorylation and glycolysis. Results Six metabolites were significantly less abundant, and two were significantly higher in abundance in HIV+ individuals compared to healthy donors. One of the HIV-upregulated metabolites, N-Acetyl-L-Alanine (ALA), inhibits pro-inflammatory cytokine IFN-□ production by NK cells of LTBI+ individuals. ALA inhibits glycolysis of LTBI+ individuals' NK cells in response to Mtb . Conclusions Our findings demonstrate that HIV infection enhances plasma ALA levels to inhibit NK cell-mediated immune responses to Mtb infection, offering a new understanding of the HIV- Mtb interaction and providing the implication of nutrition intervention and therapy for HIV- Mtb co-infected patients.
Collapse
|