1
|
de Brito CB, do Nascimento Arifa RD, de Oliveira Bezerra R, Dias Igídio CE, de Amorim-Santos BM, de Menezes Santos ACP, Barbosa LM, Barbosa JPP, Cassiano LMG, Kohlhoff M, Fagundes M, Batista RRÁ, Queiroz-Junior CM, Saliba AM, Raposo JDA, Braga FC, Coimbra RS, Teixeira MM, Fagundes CT, Souza DG. Antibiotic-Induced Dysbiosis of the Gut Microbiota Shifts Host Tryptophan Metabolism and Increases the Susceptibility of Mice to Pulmonary Infection With Pseudomonas aeruginosa. Immunology 2025. [PMID: 40387573 DOI: 10.1111/imm.13932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium that mainly infects those who have previously been treated with antibiotics. We hypothesised that antibiotic treatment disrupts tryptophan metabolism, leading to increased susceptibility to P. aeruginosa infection. Our results showed that mice receiving antibiotics exhibited intestinal dysbiosis with alterations in host tryptophan metabolism, a higher mortality rate and a higher bacterial load compared to eubiotic mice. In the lungs of the dysbiotic mice, there was an increase in IDO1 (Indoleamine 2,3-dioxygenase 1) activity and an accumulation of kynurenine after infection, and IDO1-/- mice were resistant to infection after induction of dysbiosis. Importantly, dysbiosis led to increased expression and activation of AHR (Aryl Hydrocarbon Receptor) in an IDO1-dependent manner. Blocking AHR activation in dysbiotic mice resulted in a lower bacterial load. Our data showed that increased AHR activation by kynurenine was associated with decreased phagocytosis of P. aeruginosa by macrophages and neutrophils. In conclusion, our results indicate that dysbiosis resulting from prolonged antimicrobial treatment alters tryptophan metabolism, leading to activation of the IDO1-AHR axis and increasing susceptibility to P. aeruginosa infection. Furthermore, these data suggest that IDO1 or AHR are potential host targets for the prevention of opportunistic infections in patients undergoing antimicrobial therapy.
Collapse
Affiliation(s)
- Camila Bernardo de Brito
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Raquel Duque do Nascimento Arifa
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rafael de Oliveira Bezerra
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlos Eduardo Dias Igídio
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bárbara Maria de Amorim-Santos
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anna Clara Paiva de Menezes Santos
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Larissa Mendes Barbosa
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - João Paulo Pezzini Barbosa
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Markus Kohlhoff
- Instituto René Rachou (IRR) - FIOCRUZ. FIOCRUZ MG, Belo Horizonte, MG, Brazil
| | - Micheli Fagundes
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rafaela Ribeiro Álvares Batista
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alessandra M Saliba
- Departamento de Microbiologia e Imunologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Divina Almeida Raposo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernão Castro Braga
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Mauro Martins Teixeira
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caio Tavares Fagundes
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle G Souza
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
2
|
Pei T, Li W, Zhou Z, Zhang Q, Yu G, Yin S, Chen H, Tang J. The relationship between tryptophan metabolism and gut microbiota: Interaction mechanism and potential effects in infection treatment. Microbiol Res 2025; 298:128211. [PMID: 40393170 DOI: 10.1016/j.micres.2025.128211] [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: 02/19/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
Human health is influenced by the gut microbiota, particularly in aspects of host immune homeostasis and intestinal immune response. Tryptophan (Trp) not only acts as a nutrient enhancer but also plays a critical role in the balance between host immune tolerance and gut microbiota maintenance. Both endogenous and bacterial metabolites of Trp, exert significant effects on gut microbial composition, microbial metabolism, the host immunity and the host-microbiome interface. Trp metabolites regulate host immunity by activating aryl hydrocarbon receptor (AhR), thereby contributing to immune homeostasis. Among Trp metabolites, AhR ligands include endogenous metabolites (such as kynurenine), and bacterial metabolites (such as indole and its derivatives). Here, we present a comprehensive analysis of the relationships between Trp metabolism and 14 key microbiota, encompassing fungi (e.g., Candida albicans, Aspergillus), bacteria (e.g., Ruminococcus gnavus, Bacteroides, Prevotella copri, Clostridium difficile, Escherichia coli, lactobacilli, Mycobacterium tuberculosis, Pseudomonas aeruginosa, Staphylococcus aureus, Helicobacter. Pylori), and viruses (e.g., SARS-CoV-2, influenza virus). This review clarifies how the gut microbiota regulates Trp metabolism and uncovers the underlying mechanisms of these interactions. And increased mechanistic insight into how the microbiota modulate the host immune system through Trp metabolism may allow for the identification of innovative therapies that are specifically designed to target Trp absorption, Trp metabolites, the gut microbiota, or interactions between Trp and gut microbiota to treat both intestinal and extra-intestinal inflammation as well as microbial infections.
Collapse
Affiliation(s)
- Tongchao Pei
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Wenweiran Li
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Ziyang Zhou
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Qinyu Zhang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Guohong Yu
- Department of Emergency Medicine, Baoshan Second People's Hospital, Baoshan College of Traditional Chinese Medicine, Baoshan 678000, China
| | - Sokun Yin
- Department of Emergency Medicine, Luoping County People's Hospital, Qujing 655800, China
| | - Hui Chen
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China.
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China.
| |
Collapse
|
3
|
Rangaraj S, Agarwal A, Banerjee S. Bird's Eye View on Mycobacterium tuberculosis-HIV Coinfection: Understanding the Molecular Synergism, Challenges, and New Approaches to Therapeutics. ACS Infect Dis 2025; 11:1042-1063. [PMID: 40229972 DOI: 10.1021/acsinfecdis.4c00870] [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: 04/16/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M.tb), is the most common secondary infection in the Human Immunodeficiency Virus (HIV) infected population, accounting for more than one-fourth of deaths in people living with HIV (PLWH). Reciprocally, HIV infection increases the susceptibility to primary TB or reactivation of latent TB by several folds. The synergistic interactions between M.tb and HIV not only potentiate their deleterious impact but also complicate the clinical management of both the diseases. M.tb-HIV coinfected patients have a high risk of failure of accurate diagnosis, treatment inefficiency for both TB and HIV, concurrent nontuberculous mycobacterial infections, other comorbidities such as diabetes mellitus, severe cytotoxicity due to drug overburden, and immune reconstitution inflammatory syndrome (IRIS). The need of the hour is to understand M.tb-HIV coinfection biology and their collective impact on the host immunocompetence and to think of out-of-the-box treatment perspectives, including host-directed therapy under the rising view of homeostatic medicines. This review aims to highlight the molecular players, both from the pathogens and host, that facilitate the synergistic interactions and host-associated proteins/enzymes regulating immunometabolism, underlining potential targets for designing and screening chemical inhibitors to reduce the burden of both pathogens concomitantly during M.tb-HIV coinfection. To appreciate the necessity of revisiting therapeutic approaches and research priorities, we provide a glimpse of anti-TB and antiretroviral drug-drug interactions, project the gaps in our understanding of coinfection biology, and also enlist some key research initiatives that will help us deal with the synergistic epidemic of M.tb-HIV coinfection.
Collapse
Affiliation(s)
- Siranjeevi Rangaraj
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Anushka Agarwal
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Sharmistha Banerjee
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| |
Collapse
|
4
|
Collins JM, Kipiani M, Jin Y, Sharma AA, Tomalka JA, Avaliani T, Gujabidze M, Bakuradze T, Sabanadze S, Avaliani Z, Blumberg HM, Benkeser D, Jones DP, Peloquin C, Kempker RR. Pharmacometabolomics in TB meningitis-Understanding the pharmacokinetic, metabolic, and immune factors associated with anti-TB drug concentrations in cerebrospinal fluid. PLoS One 2025; 20:e0315999. [PMID: 40029856 PMCID: PMC11875335 DOI: 10.1371/journal.pone.0315999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/04/2024] [Indexed: 03/06/2025] Open
Abstract
Poor penetration of many anti-tuberculosis (TB) antibiotics into the central nervous system (CNS) is thought to be a major driver of morbidity and mortality in TB meningitis (TBM). While the amount of a particular drug that crosses into the cerebrospinal fluid (CSF) varies from person to person, little is known about the host factors associated with interindividual differences in CSF concentrations of anti-TB drugs. In patients diagnosed with TBM from the country of Georgia (n = 17), we investigate the association between CSF concentrations of anti-TB antibiotics and multiple host factors including serum drug concentrations and CSF concentrations of metabolites and cytokines. We found > 2-fold differences in CSF concentrations of anti-TB antibiotics from person to person for all drugs tested including cycloserine, ethambutol, imipenem, isoniazid, levofloxacin, linezolid, moxifloxacin, pyrazinamide, and rifampin. While serum drug concentrations explained over 30% of the variation in CSF drug concentrations for cycloserine, isoniazid, linezolid, and pyrazinamide (adjusted R2 ≥ 0.3, p < 0.001 for all), there was no significant association between serum concentrations of imipenem and ethambutol and their respective CSF concentrations. CSF concentrations of carnitines were significantly associated with concentrations of ethambutol and imipenem (q < 0.05), and imipenem was the only antibiotic significantly associated with CSF cytokine concentrations. These results indicate that there is high interindividual variability in CSF drug concentrations in patients treated for TBM, which is only partially explained by differences in serum drug concentrations. With the exception of imipenem, there was no association between CSF drug concentrations and concentrations of cytokines and chemokines.
Collapse
Affiliation(s)
- Jeffrey M. Collins
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Maia Kipiani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
- The University of Georgia, Tbilisi, Georgia
- David Tvildiani Medical University, The University of Georgia, Tbilisi, Georgia
| | - Yutong Jin
- Department of Biostatistics, Rollins School of Public Health of Emory University, Atlanta, Georgia, United States of America
| | - Ashish A. Sharma
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jeffrey A. Tomalka
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Teona Avaliani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Mariam Gujabidze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Tinatin Bakuradze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Shorena Sabanadze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Zaza Avaliani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
- European University, Tbilisi, Georgia
| | - Henry M. Blumberg
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Departments of Epidemiology and Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - David Benkeser
- Department of Biostatistics, Rollins School of Public Health of Emory University, Atlanta, Georgia, United States of America
| | - Dean P. Jones
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Charles Peloquin
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, United States of America
| | - Russell R. Kempker
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
5
|
McCaffrey EF, Delmastro AC, Fitzhugh I, Ranek JS, Douglas S, Peters JM, Fullaway CC, Bosse M, Liu CC, Gillen C, Greenwald NF, Anzick S, Martens C, Winfree S, Bai Y, Sowers C, Goldston M, Kong A, Boonrat P, Bigbee CL, Venugopalan R, Maiello P, Klein E, Rodgers MA, Scanga CA, Lin PL, Kirschner D, Fortune S, Bryson BD, Butler JR, Mattila JT, Flynn JL, Angelo M. The immunometabolic topography of tuberculosis granulomas governs cellular organization and bacterial control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638923. [PMID: 40027668 PMCID: PMC11870603 DOI: 10.1101/2025.02.18.638923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Despite being heavily infiltrated by immune cells, tuberculosis (TB) granulomas often subvert the host response to Mycobacterium tuberculosis (Mtb) infection and support bacterial persistence. We previously discovered that human TB granulomas are enriched for immunosuppressive factors typically associated with tumor-immune evasion, raising the intriguing possibility that they promote tolerance to infection. In this study, our goal was to identify the prime drivers for establishing this tolerogenic niche and to determine if the magnitude of this response correlates with bacterial persistence. To do this, we conducted a multimodal spatial analysis of 52 granulomas from 16 non-human primates (NHP) who were infected with low dose Mtb for 9-12 weeks. Notably, each granuloma's bacterial burden was individually quantified allowing us to directly ask how granuloma spatial structure and function relate to infection control. We found that a universal feature of TB granulomas was partitioning of the myeloid core into two distinct metabolic environments, one of which is hypoxic. This hypoxic environment associated with pathologic immune cell states, dysfunctional cellular organization of the granuloma, and a near-complete blockade of lymphocyte infiltration that would be required for a successful host response. The extent of these hypoxia-associated features correlated with worsened bacterial burden. We conclude that hypoxia governs immune cell state and organization within granulomas and is a potent driver of subverted immunity during TB.
Collapse
Affiliation(s)
- Erin F. McCaffrey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
- Spatial Immunology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD
| | - Alea C. Delmastro
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Isobel Fitzhugh
- The Department of Biomedical Sciences and Technology, AdventHealth University, Orlando, FL
| | - Jolene S. Ranek
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Sarah Douglas
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Joshua M. Peters
- Department of Biological Engineering, MIT, Cambridge, MA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, MA
| | | | - Marc Bosse
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Candace C. Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Craig Gillen
- The Department of Biomedical Sciences and Technology, AdventHealth University, Orlando, FL
| | - Noah F. Greenwald
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Sarah Anzick
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT
| | - Craig Martens
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT
| | - Seth Winfree
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, MT
| | - Yunhao Bai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Cameron Sowers
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Mako Goldston
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Alex Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Potchara Boonrat
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Carolyn L. Bigbee
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Roopa Venugopalan
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Edwin Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark A. Rodgers
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Philana Ling Lin
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Pediatrics, Division of Infectious Disease, Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Denise Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Sarah Fortune
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, MA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Bryan D. Bryson
- Department of Biological Engineering, MIT, Cambridge, MA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, MA
| | - J. Russell Butler
- The Department of Biomedical Sciences and Technology, AdventHealth University, Orlando, FL
| | - Joshua T. Mattila
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
6
|
Liu X, Yang M, Xu P, Du M, Li S, Shi J, Li Q, Yuan J, Pang Y. Kynurenine-AhR reduces T-cell infiltration and induces a delayed T-cell immune response by suppressing the STAT1-CXCL9/CXCL10 axis in tuberculosis. Cell Mol Immunol 2024; 21:1426-1440. [PMID: 39438693 PMCID: PMC11607402 DOI: 10.1038/s41423-024-01230-1] [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: 05/10/2024] [Revised: 09/20/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is a critical global health issue that is complicated by the ability of the pathogen to delay the host's T-cell immune response. This delay in T-cell recruitment to the site of infection is a pivotal survival strategy for Mtb, allowing it to establish a persistent chronic infection. To investigate the underlying mechanisms, this study focused on Mtb's exploitation of host tryptophan metabolism. Mtb upregulates indoleamine 2,3-dioxygenase 1 (IDO1) in inflammatory macrophages, thereby increasing kynurenine (Kyn) production. Kyn then activates the aryl hydrocarbon receptor (AhR), leading to the upregulation of suppressor of cytokine signaling 3 and subsequent inhibition of the JAK-STAT1 signaling pathway. This results in reduced secretion of the chemokines CXCL9 and CXCL10, which are crucial for T-cell recruitment to the lungs. Supported by in vivo mouse models, our findings reveal that disrupting this pathway through AhR knockout significantly enhances T-cell infiltration and activity, thereby undermining Mtb-induced immunosuppression. In contrast, additional Kyn injection obviously inhibited T-cell infiltration and activity. These results highlight potential therapeutic targets of AhR and IDO1, offering new avenues for enhancing the host immune response against tuberculosis and guiding future vaccine development efforts.
Collapse
Affiliation(s)
- Xin Liu
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Mengjie Yang
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Ping Xu
- The Affiliated Infectious Diseases Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Mingwei Du
- The Affiliated Infectious Diseases Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Shanshan Li
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Jin Shi
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Qiang Li
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Jinfeng Yuan
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Yu Pang
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| |
Collapse
|
7
|
Liu R, Zhang F, Li S, Liu Q, Pang Y, Li L. Regulation of ROS metabolism in macrophage via xanthine oxidase is associated with disease progression in pulmonary tuberculosis. Metabolomics 2024; 20:127. [PMID: 39520502 DOI: 10.1007/s11306-024-02194-z] [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: 08/14/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Pulmonary tuberculosis (PTB) exacerbation can lead to respiratory failure, multi-organ failure, and symptoms related to central nervous system diseases. The purpose of this study is to screen biomarkers and metabolic pathways that can predict the progression of PTB, and to verify the role of the metabolic enzyme xanthine oxidase (XO) in the progression of PTB. METHODS To explore the biomarkers and mechanisms underlying the progression of PTB, plasma metabolomics sequencing was conducted on patients with severe PTB, non-severe PTB, and healthy individuals. Screening differential metabolites and metabolic pathways that can predict the progression of PTB, and verifying the function and mechanism of action of XO through experiments. RESULTS The purine metabolism, sphingolipid metabolism, and amino acid metabolism between the three groups differ. In patients with severe PTB, the levels of xanthosine and hypoxanthine are increased, while the levels of D-tryptophan, dihydroceramide and uric acid are decreased. Inhibition of XO activity has been observed to reduce the levels of tumor necrosis factor (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6), as well as to suppress the production of reactive oxygen species (ROS) and the activation of the NF-κB pathway, while also promoting the growth of MTB within cells. CONCLUSION D-tryptophan, xanthosine, and dihydroceramide can be utilized as biomarkers for progression of PTB, assisting in the evaluation of disease progression, and XO stands out as a potential therapeutic target for impeding the progression of PTB.
Collapse
Affiliation(s)
- Ruichao Liu
- Department of Bacteriology and Immunology, Beijing Tuberculosis & Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, 101149, P.R. China
| | - Fuzhen Zhang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P.R. China
| | - Shanshan Li
- Department of Bacteriology and Immunology, Beijing Tuberculosis & Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, 101149, P.R. China
| | - Qiuyue Liu
- Department of Intensive Care Unit, Beijing Tuberculosis & Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, 101149, P.R. China.
| | - Yu Pang
- Department of Bacteriology and Immunology, Beijing Tuberculosis & Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, 101149, P.R. China.
| | - Liang Li
- Department of Bacteriology and Immunology, Beijing Tuberculosis & Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, 101149, P.R. China.
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, P.R. China.
| |
Collapse
|
8
|
Chakhaia T, Blumberg HM, Kempker RR, Luo R, Dzidzikashvili N, Chincharauli M, Tukvadze N, Avaliani Z, Stauber C, Magee MJ. Lack of weight gain and increased mortality during and after treatment among adults with drug-resistant tuberculosis in Georgia, 2009-2020. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.05.24311499. [PMID: 39148852 PMCID: PMC11326334 DOI: 10.1101/2024.08.05.24311499] [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: 08/17/2024]
Abstract
Background While low body mass index (BMI) is associated with poor tuberculosis (TB) treatment outcomes, the impact of weight gain during TB treatment is unclear. To address this knowledge gap, we assessed if lack of weight gain is associated with all-cause mortality during and after TB treatment. Methods We conducted a retrospective cohort study among adults with newly diagnosed multi- or extensively drug-resistant (M/XDR) pulmonary TB in Georgia between 2009-2020. The exposure was a change in BMI during the first 3-6 months of TB treatment. All-cause mortality during and after TB treatment was assessed using the National Death Registry. We used competing-risk Cox proportional hazard models to estimate adjusted hazard ratios (aHR) between BMI change and all-cause mortality. Results Among 720 adult participants, 21% had low BMI (<18.5 kg/m2) at treatment initiation and 9% died either during (n=16) or after treatment (n=50). During the first 3-6 months of TB treatment, 17% lost weight and 14% had no weight change. Among 479 adults with normal baseline BMI ( ≥18.5-24.9 kg/m2), weight loss was associated with an increased risk of death during TB treatment (aHR=5.25; 95%CI: 1.31-21.10). Among 149 adults with a low baseline BMI, no change in BMI was associated with increased post-TB treatment mortality (aHR=4.99; 95%CI: 1.25-19.94). Conclusions Weight loss during TB treatment (among those with normal baseline BMI) or no weight gain (among those with low baseline BMI) was associated with increased rates of all-cause mortality. Our findings suggest that scaling up weight management interventions among those with M/XDR TB may be beneficial.
Collapse
Affiliation(s)
- Tsira Chakhaia
- Georgia State University, Atlanta, GA, USA
- National Center for Tuberculosis and Lung Disease, Tbilisi, Georgia
| | | | | | - Ruiyan Luo
- Georgia State University, Atlanta, GA, USA
| | | | | | - Nestan Tukvadze
- National Center for Tuberculosis and Lung Disease, Tbilisi, Georgia
- Swiss Tropical and Public Health Institute
| | - Zaza Avaliani
- National Center for Tuberculosis and Lung Disease, Tbilisi, Georgia
| | | | | |
Collapse
|
9
|
Singh B, Sharan R, Ravichandran G, Escobedo R, Shivanna V, Dick EJ, Hall-Ursone S, Arora G, Alvarez X, Singh DK, Kaushal D, Mehra S. Indoleamine-2,3-dioxygenase inhibition improves immunity and is safe for concurrent use with cART during Mtb/SIV coinfection. JCI Insight 2024; 9:e179317. [PMID: 39114981 PMCID: PMC11383603 DOI: 10.1172/jci.insight.179317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/26/2024] [Indexed: 09/13/2024] Open
Abstract
Chronic immune activation promotes tuberculosis (TB) reactivation in the macaque Mycobacterium tuberculosis (M. tuberculosis)/SIV coinfection model. Initiating combinatorial antiretroviral therapy (cART) early lowers the risk of TB reactivation, but immune activation persists. Studies of host-directed therapeutics (HDTs) that mitigate immune activation are, therefore, required. Indoleamine 2,3, dioxygenase (IDO), a potent immunosuppressor, is one of the most abundantly induced proteins in NHP and human TB granulomas. Inhibition of IDO improves immune responses in the lung, leading to better control of TB, including adjunctive to TB chemotherapy. The IDO inhibitor D-1 methyl tryptophan (D1MT) is, therefore, a bona fide TB HDT candidate. Since HDTs against TB are likely to be deployed in an HIV coinfection setting, we studied the effect of IDO inhibition in M. tuberculosis/SIV coinfection, adjunctive to cART. D1MT is safe in this setting, does not interfere with viral suppression, and improves the quality of CD4+ and CD8+ T cell responses, including reconstitution, activation and M. tuberculosis-specific cytokine production, and access of CD8+ T cells to the lung granulomas; it reduces granuloma size and necrosis, type I IFN expression, and the recruitment of inflammatory IDO+ interstitial macrophages (IMs). Thus, trials evaluating the potential of IDO inhibition as HDT in the setting of cART in M. tuberculosis/HIV coinfected individuals are warranted.
Collapse
|
10
|
Maulina N, Hayati Z, Hasballah K, Zulkarnain Z. Tryptophan and Its Derived Metabolites as Biomarkers for Tuberculosis Disease: A Systematic Review. IRANIAN BIOMEDICAL JOURNAL 2024; 28:140-7. [PMID: 39034495 PMCID: PMC11444479 DOI: 10.61186/ibj.4174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Feasible diagnostic assays are required to detect new tuberculosis (TB) cases and monitor treatment. This study aimed to evaluate evidence on tryptophan (Trp) and its metabolites as proposed biomarkers for TB. Through specific keyword searches, we identified 170 relevant literature sources and included seven publications (from 2013 to 2023). The biomarker used in these studies were indoleamine 2, 3-dioxygenase (IDO) activity, IDO-1 gene expression, and plasma IDO protein, measured using ELISA, liquid chromatography-mass spectrometry, ultraperformance liquid chromatography mass spectrometry, and transcriptional profiling. The studies encompassed a pediatric case-control and six studies involving adults, pregnant women with TB-HIV, and individuals with multidrug-resistant tuberculosis, active TB, and latent TB. The assessment of IDO activity and IDO protein level demonstrated promising performance in distinguishing active TB from controls and in evaluating treatment failure and recurrent cases to controls. Trp and its metabolites fulfilled nearly all of target product profile criteria for detecting active TB. This study highlights the potential of utilizing host Trp and its metabolites as non-sputum-based biomarker for TB infection.
Collapse
Affiliation(s)
- Novi Maulina
- Doctorate Student of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, 23116, Indonesia
- Microbiology Department, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, 23116, Indonesia
| | - Zinatul Hayati
- Microbiology Department, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, 23116, Indonesia
| | - Kartini Hasballah
- Pharmacology Department, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, 23116, Indonesia
| | - Zulkarnain Zulkarnain
- Physiology Department, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, 23116, Indonesia
| |
Collapse
|
11
|
Li M, Zhang D, Yang Q, Zhao Z, Zhang C, Zhou Y, Bai Y, Chen L, Tang X, Liu C, Zhou J, Chen X, Ying B. Longitudinal metabolomics of human plasma reveal metabolic dynamics and predictive markers of antituberculosis drug-induced liver injury. Respir Res 2024; 25:254. [PMID: 38907347 PMCID: PMC11193241 DOI: 10.1186/s12931-024-02837-8] [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: 11/04/2023] [Accepted: 05/04/2024] [Indexed: 06/23/2024] Open
Abstract
Tuberculosis (TB) remains the second leading cause of death from a single infectious agent and long-term medication could lead to antituberculosis drug-induced liver injury (ATB-DILI). We established a prospective longitudinal cohort of ATB-DILI with multiple timepoint blood sampling and used untargeted metabolomics to analyze the metabolic profiles of 107 plasma samples from healthy controls and newly diagnosed TB patients who either developed ATB-DILI within 2 months of anti-TB treatment (ATB-DILI subjects) or completed their treatment without any adverse drug reaction (ATB-Ctrl subjects). The untargeted metabolome revealed that 77 metabolites (of 895 total) were significantly changed with ATB-DILI progression. Among them, levels of multiple fatty acids and bile acids significantly increased over time in ATB-DILI subjects. Meanwhile, metabolites of the same class were highly correlated with each other and pathway analysis indicated both fatty acids metabolism and bile acids metabolism were up-regulated with ATB-DILI progression. The targeted metabolome further validated that 5 fatty acids had prediction capability at the early stage of the disease and 6 bile acids had a better diagnostic performance when ATB-DILI occurred. These findings provide evidence indicating that fatty acids metabolism and bile acids metabolism play a vital role during ATB-DILI progression. Our report adds a dynamic perspective better to understand the pathological process of ATB-DILI in clinical settings.
Collapse
Affiliation(s)
- Mengjiao Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Zhang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qingxin Yang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenzhen Zhao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chunying Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yanbing Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yangjuan Bai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Chen
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyan Tang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Cuihua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Xuerong Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Kreimendahl S, Pernas L. Metabolic immunity against microbes. Trends Cell Biol 2024; 34:496-508. [PMID: 38030541 DOI: 10.1016/j.tcb.2023.10.013] [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: 06/09/2023] [Revised: 10/11/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
Pathogens, including viruses, bacteria, fungi, and parasites, remodel the metabolism of their host to acquire the nutrients they need to proliferate. Thus, host cells are often perceived as mere exploitable nutrient pools during infection. Mounting reports challenge this perception and instead suggest that host cells can actively reprogram their metabolism to the detriment of the microbial invader. In this review, we present metabolic mechanisms that host cells use to defend against pathogens. We highlight the contribution of domesticated microbes to host defenses and discuss examples of host-pathogen arms races that are derived from metabolic conflict.
Collapse
Affiliation(s)
| | - Lena Pernas
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
13
|
Ogongo P, Tran A, Marzan F, Gingrich D, Krone M, Aweeka F, Lindestam Arlehamn CS, Martin JN, Deeks SG, Hunt PW, Ernst JD. High-parameter phenotypic characterization reveals a subset of human Th17 cells that preferentially produce IL-17 against M. tuberculosis antigen. Front Immunol 2024; 15:1378040. [PMID: 38698866 PMCID: PMC11064812 DOI: 10.3389/fimmu.2024.1378040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Background Interleukin-17-producing CD4 T cells contribute to the control of Mycobacterium tuberculosis (Mtb) infection in humans; whether infection with human immunodeficiency virus (HIV) disproportionately affects distinct Th17-cell subsets that respond to Mtb is incompletely defined. Methods We performed high-definition characterization of circulating Mtb-specific Th17 cells by spectral flow cytometry in people with latent TB and treated HIV (HIV-ART). We also measured kynurenine pathway activity by liquid chromatography-mass spectrometry (LC/MS) on plasma and tested the hypothesis that tryptophan catabolism influences Th17-cell frequencies in this context. Results We identified two subsets of Th17 cells: subset 1 defined as CD4+Vα7.2-CD161+CD26+and subset 2 defined as CD4+Vα7.2-CCR6+CXCR3-cells of which subset 1 was significantly reduced in latent tuberculosis infection (LTBI) with HIV-ART, yet Mtb-responsive IL-17-producing CD4 T cells were preserved; we found that IL-17-producing CD4 T cells dominate the response to Mtb antigen but not cytomegalovirus (CMV) antigen or staphylococcal enterotoxin B (SEB), and tryptophan catabolism negatively correlates with both subset 1 and subset 2 Th17-cell frequencies. Conclusions We found differential effects of ART-suppressed HIV on distinct subsets of Th17 cells, that IL-17-producing CD4 T cells dominate responses to Mtb but not CMV antigen or SEB, and that kynurenine pathway activity is associated with decreases of circulating Th17 cells that may contribute to tuberculosis immunity.
Collapse
Affiliation(s)
- Paul Ogongo
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
| | - Anthony Tran
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Florence Marzan
- Drug Research Unit, Department of Clinical Pharmacy, School of Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - David Gingrich
- Drug Research Unit, Department of Clinical Pharmacy, School of Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Melissa Krone
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Francesca Aweeka
- Drug Research Unit, Department of Clinical Pharmacy, School of Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | | | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Peter W. Hunt
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Joel D. Ernst
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
14
|
Fairley JK, Ferreira JA, Fraga LAO, Lyon S, Valadão Cardoso TM, Boson VC, Madureira Nunes AC, Medeiros Cinha EH, de Oliveira LBP, Magueta Silva EB, Marçal PHF, Branco AC, Grossi MAF, Jones DP, Ziegler TR, Collins JM. High-Resolution Plasma Metabolomics Identifies Alterations in Fatty Acid, Energy, and Micronutrient Metabolism in Adults Across the Leprosy Spectrum. J Infect Dis 2024; 229:1189-1199. [PMID: 37740551 PMCID: PMC11011203 DOI: 10.1093/infdis/jiad410] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND High-resolution metabolomics (HRM) is an innovative tool to study challenging infectious diseases like leprosy, where the pathogen cannot be grown with standard methods. Here, we use HRM to better understand associations between disease manifestations, nutrition, and host metabolism. METHODS From 2018 to 2019, adults with leprosy and controls were recruited in Minas Gerais, Brazil. Plasma metabolites were detected using an established HRM workflow and characterized by accurate mass, mass to charge ratio m/z and retention time. The mummichog informatics package compared metabolic pathways between cases and controls and between multibacillary (MB) and paucibacillary (PB) leprosy. Additionally, select individual metabolites were quantified and compared. RESULTS Thirty-nine cases (62% MB and 38% PB) and 25 controls were enrolled. We found differences (P < .05) in several metabolic pathways, including fatty acid metabolism, carnitine shuttle, retinol, vitamin D3, and C-21 steroid metabolism, between cases and controls with lower retinol and associated metabolites in cases. Between MB and PB, leukotrienes, prostaglandins, tryptophan, and cortisol were all found to be lower in MB (P < .05). DISCUSSION Metabolites associated with several nutrient-related metabolic pathways appeared differentially regulated in leprosy, especially MB versus PB. This pilot study demonstrates the metabolic interdependency of these pathways, which may play a role in the pathophysiology of disease.
Collapse
Affiliation(s)
- Jessica K Fairley
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - José A Ferreira
- Faculdade da Saúde e Écologia Humana, FASEH, Vespasiano, Minas Gerais, Brazil
| | - Lucia A O Fraga
- Department of Health Sciences, Universidade Federal de Juiz de Fora, Governador Valadares, Minas Gerais, Brazil
| | - Sandra Lyon
- Faculdade da Saúde e Écologia Humana, FASEH, Vespasiano, Minas Gerais, Brazil
- Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Victor Campos Boson
- Faculdade da Saúde e Écologia Humana, FASEH, Vespasiano, Minas Gerais, Brazil
| | | | - Eloisa H Medeiros Cinha
- Department of Health Sciences, Universidade Federal de Juiz de Fora, Governador Valadares, Minas Gerais, Brazil
| | - Lorena B P de Oliveira
- Department of Health Sciences, Universidade Federal de Juiz de Fora, Governador Valadares, Minas Gerais, Brazil
| | - Erica B Magueta Silva
- Department of Health Sciences, Universidade Federal de Juiz de Fora, Governador Valadares, Minas Gerais, Brazil
| | - Pedro H F Marçal
- Department of Health Sciences, Universidade Federal de Juiz de Fora, Governador Valadares, Minas Gerais, Brazil
| | - Alexandre C Branco
- Centro de Referência em Doenças Endêmicas e Programs Especiais, Governador Valadares, Minas Gerais, Brazil
| | | | - Dean P Jones
- Division of Pulmonary, Critical Care, and Allergy, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Thomas R Ziegler
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jeffrey M Collins
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Gatechompol S, Lutter R, Vaz FM, Ubolyam S, Avihingsanon A, Kerr SJ, van Leth F, Cobelens F. The plasma kynurenine-to-tryptophan ratio as a biomarker of tuberculosis disease in people living with HIV on antiretroviral therapy: an exploratory nested case-control study. BMC Infect Dis 2024; 24:372. [PMID: 38565993 PMCID: PMC10988902 DOI: 10.1186/s12879-024-09258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Non-sputum-based tests are needed to predict or diagnose tuberculosis (TB) disease in people living with HIV (PWH). The enzyme indoleamine 2, 3-dioxygenase-1 (IDO1) is expressed in tuberculoid granuloma and catabolizes tryptophan (Trp) to kynurenine (Kyn). IDO1 activity compromises innate and adaptive immune responses, promoting mycobacterial survival. The plasma Kyn-to-Trp (K/T) ratio is a potential TB diagnostic and/or predictive biomarker in PWH on long-term antiretroviral therapy (ART). METHODS We compared plasma K/T ratios in samples from PWH, who were followed up prospectively and developed TB disease after ART initiation. Controls were matched for age and duration of ART. Kyn and Trp were measured at 3 timepoints; at TB diagnosis, 6 months before TB diagnosis and 6 months after TB diagnosis, using ultra performance liquid chromatography combined with mass spectrometry. RESULTS The K/T ratios were higher for patients with TB disease at time of diagnosis (median, 0.086; IQR, 0.069-0.123) compared to controls (0.055; IQR 0.045-0.064; p = 0.006), but not before or after TB diagnosis. K/T ratios significantly declined after successful TB treatment, but increased upon treatment failure. The K/T ratios showed a parabolic correlation with CD4 cell counts in participants with TB (p = 0.005), but there was no correlation in controls. CONCLUSIONS The plasma K/T ratio helped identify TB disease and may serve as an adjunctive biomarker for for monitoring TB treatment in PWH. Validation studies to ascertain these findings and evaluate the optimum cut-off for diagnosis of TB disease in PWH should be undertaken in well-designed prospective cohorts. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT00411983.
Collapse
Grants
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Department of Experimental Immunology, Amsterdam UMC and Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, The Netherlands
- Department of Core Facility Metabolomics, Amsterdam UMC, Amsterdam, The Netherlands.
- Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam Public Health research institute, Amsterdam, The Netherlands
- Department of Global Health and Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers location University of Amsterdam, Amsterdam, The Netherlands.
Collapse
Affiliation(s)
- Sivaporn Gatechompol
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand.
- Center of Excellence in Tuberculosis, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Department of Global Health and Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers Location University of Amsterdam, Amsterdam, The Netherlands.
| | - René Lutter
- Department of Experimental Immunology, Amsterdam UMC and Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pulmonary Diseases, Amsterdam UMC, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Department of Core Facility Metabolomics, Amsterdam UMC, Amsterdam, The Netherlands
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sasiwimol Ubolyam
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Center of Excellence in Tuberculosis, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Anchalee Avihingsanon
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Center of Excellence in Tuberculosis, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Stephen J Kerr
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Biostatistics Excellence Centre, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Frank van Leth
- Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Frank Cobelens
- Department of Global Health and Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers Location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Zhang K, Mishra A, Jagannath C. New insight into arginine and tryptophan metabolism in macrophage activation during tuberculosis. Front Immunol 2024; 15:1363938. [PMID: 38605962 PMCID: PMC11008464 DOI: 10.3389/fimmu.2024.1363938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Arginine and tryptophan are pivotal in orchestrating cytokine-driven macrophage polarization and immune activation. Specifically, interferon-gamma (IFN-γ) stimulates inducible nitric oxide synthase (iNOS) expression), leading to the conversion of arginine into citrulline and nitric oxide (NO), while Interleukin-4 (IL4) promotes arginase activation, shifting arginine metabolism toward ornithine. Concomitantly, IFN-γ triggers indoleamine 2,3-dioxygenase 1 (IDO1) and Interleukin-4 induced 1 (IL4i1), resulting in the conversion of tryptophan into kynurenine and indole-3-pyruvic acid. These metabolic pathways are tightly regulated by NAD+-dependent sirtuin proteins, with Sirt2 and Sirt5 playing integral roles. In this review, we present novel insights that augment our understanding of the metabolic pathways of arginine and tryptophan following Mycobacterium tuberculosis infection, particularly their relevance in macrophage responses. Additionally, we discuss arginine methylation and demethylation and the role of Sirt2 and Sirt5 in regulating tryptophan metabolism and arginine metabolism, potentially driving macrophage polarization.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Abhishek Mishra
- 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
|
17
|
Pamart G, Gosset P, Le Rouzic O, Pichavant M, Poulain-Godefroy O. Kynurenine Pathway in Respiratory Diseases. Int J Tryptophan Res 2024; 17:11786469241232871. [PMID: 38495475 PMCID: PMC10943758 DOI: 10.1177/11786469241232871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/28/2024] [Indexed: 03/19/2024] Open
Abstract
The kynurenine pathway is the primary route for tryptophan catabolism and has received increasing attention as its association with inflammation and the immune system has become more apparent. This review provides a broad overview of the kynurenine pathway in respiratory diseases, from the initial observations to the characterization of the different cell types involved in the synthesis of kynurenine metabolites and the underlying immunoregulatory mechanisms. With a focus on respiratory infections, the various attempts to characterize the kynurenine/tryptophan (K/T) ratio as an inflammatory marker are reviewed. Its implication in chronic lung inflammation and its exacerbation by respiratory pathogens is also discussed. The emergence of preclinical interventional studies targeting the kynurenine pathway opens the way for the future development of new therapies.
Collapse
Affiliation(s)
- Guillaume Pamart
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Philippe Gosset
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Olivier Le Rouzic
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Muriel Pichavant
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Odile Poulain-Godefroy
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
18
|
Ogongo P, Tran A, Marzan F, Gingrich D, Krone M, Aweeka F, Lindestam Arlehamn CS, Martin JN, Deeks SG, Hunt PW, Ernst JD. High-parameter phenotypic characterization reveals a subset of human Th17 cells that preferentially produce IL17 against M. tuberculosis antigen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.06.523027. [PMID: 36711855 PMCID: PMC9881994 DOI: 10.1101/2023.01.06.523027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Interleukin 17 producing CD4 T cells contribute to the control of Mycobacterium tuberculosis (Mtb) infection in humans; whether infection with Human Immunodeficiency Virus (HIV) disproportionately affects distinct Th17 cell subsets that respond to Mtb is incompletely defined. Methods We performed high-definition characterization of circulating Mtb-specific Th17 cells by spectral flow cytometry in people with latent TB and treated HIV (HIV-ART). We also measured kynurenine pathway activity by LC/MS on plasma and tested the hypothesis that tryptophan catabolism influences Th17 cell frequencies in this context. Results We identified two subsets of Th17 cells: subset 1 defined as CD4+Vα7.2-CD161+CD26+ and subset 2 defined as CD4+Vα7.2-CCR6+CXCR3- cells of which subset 1 was significantly reduced in LTBI with HIV-ART, yet Mtb-responsive IL17-producing CD4 T cells were preserved; we found that IL17-producing CD4 T cells dominate the response to Mtb antigen but not CMV antigen or staphylococcal enterotoxin B (SEB); and tryptophan catabolism negatively correlates with both subset 1 and subset 2 Th17 cell frequencies. Conclusions We found differential effects of ART-suppressed HIV on distinct subsets of Th17 cells, that IL17-producing CD4 T cells dominate responses to Mtb but not CMV antigen or SEB, and that kynurenine pathway activity is associated with decreases of circulating Th17 cells that may contribute to tuberculosis immunity.
Collapse
Affiliation(s)
- Paul Ogongo
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
| | - Anthony Tran
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Florence Marzan
- Drug Research Unit, Department of Clinical Pharmacy, School of Pharmacy, University of California, San Francisco, CA, USA
| | - David Gingrich
- Drug Research Unit, Department of Clinical Pharmacy, School of Pharmacy, University of California, San Francisco, CA, USA
| | - Melissa Krone
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Francesca Aweeka
- Drug Research Unit, Department of Clinical Pharmacy, School of Pharmacy, University of California, San Francisco, CA, USA
| | | | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Peter W. Hunt
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Joel D. Ernst
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
19
|
Arya R, Jit BP, Kumar V, Kim JJ. Exploring the Potential of Exosomes as Biomarkers in Tuberculosis and Other Diseases. Int J Mol Sci 2024; 25:2885. [PMID: 38474139 DOI: 10.3390/ijms25052885] [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: 01/01/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Tuberculosis (TB) is a major cause of morbidity and mortality and remains an important public health issue in developing countries worldwide. The existing methods and techniques available for the diagnosis of TB are based on combinations of laboratory (chemical and biological), radiological, and clinical tests. These methods are sophisticated and laborious and have limitations in terms of sensitivity, specificity, and accuracy. Clinical settings need improved diagnostic biomarkers to accurately detect biological changes due to pathogen invasion and pharmacological responses. Exosomes are membrane-bound vesicles and mediators of intercellular signaling processes that play a significant role in the pathogenesis of various diseases, such as tuberculosis, and can act as promising biomarkers for the monitoring of TB infection. Compared to conventional biomarkers, exosome-derived biomarkers are advantageous because they are easier to detect in different biofluids, are more sensitive and specific, and may be useful in tracking patients' reactions to therapy. This review provides insights into the types of biomarkers, methods of exosome isolation, and roles of the cargo (proteins) present in exosomes isolated from patients through omics studies, such as proteomics. These findings will aid in developing new prognostic and diagnostic biomarkers and could lead to the identification of new therapeutic targets in the clinical setting.
Collapse
Affiliation(s)
- Rakesh Arya
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijay Kumar
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jong Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
20
|
Zhang SS, Yang X, Zhang WX, Zhou Y, Wei TT, Cui N, Du J, Liu W, Lu QB. Metabolic alterations in urine among the patients with severe fever with thrombocytopenia syndrome. Virol J 2024; 21:11. [PMID: 38191404 PMCID: PMC10775654 DOI: 10.1186/s12985-024-02285-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/02/2024] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND The pathogenesis of severe fever with thrombocytopenia syndrome (SFTS) remained unclear. We aimed to profile the metabolic alterations in urine of SFTS patients and provide new evidence for its pathogenesis. METHODS A case-control study was conducted in the 154th hospital in China. Totally 88 cases and 22 controls aged ≥ 18 years were enrolled. The cases were selected from laboratory-confirmed SFTS patients. The controls were selected among SFTSV-negative population. Those with diabetes, cancer, hepatitis and other sexually transmitted diseases were excluded in both groups. Fatal cases and survival cases were 1:1 matched. Inter-group differential metabolites and pathways were obtained, and the inter-group discrimination ability was evaluated. RESULTS Tryptophan metabolism and phenylalanine metabolism were the top one important metabolism pathway in differentiating the control and case groups, and the survival and fatal groups, respectively. The significant increase of differential metabolites in tryptophan metabolism, including 5-hydroxyindoleacetate (5-HIAA), L-kynurenine (KYN), 5-hydroxy-L-tryptophan (5-HTP), 3-hydroxyanthranilic acid (3-HAA), and the increase of phenylpyruvic acid and decrease of hippuric acid in phenylalanine metabolism indicated the potential metabolic alterations in SFTSV infection. The increase of 5-HIAA, KYN, 5-HTP, phenylpyruvic acid and hippuric acid were involved in the fatal progress of SFTS patients. CONCLUSIONS Tryptophan metabolism and phenylalanine metabolism might be involved in the pathogenesis of SFTSV infection. These findings provided new evidence for the pathogenesis and treatment of SFTS.
Collapse
Affiliation(s)
- Shan-Shan Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xin Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Wan-Xue Zhang
- Center for Infectious Disease and Policy Research & Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Yiguo Zhou
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China
| | - Ting-Ting Wei
- Department of Laboratorial of Science and Technology & Vaccine Research Center, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing, 100191, China
| | - Ning Cui
- Department of Infectious Diseases, The 154th Hospital, Xinyang, China
| | - Juan Du
- Department of Laboratorial of Science and Technology & Vaccine Research Center, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing, 100191, China
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Qing-Bin Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.
- Center for Infectious Disease and Policy Research & Global Health and Infectious Diseases Group, Peking University, Beijing, China.
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China.
- Department of Laboratorial of Science and Technology & Vaccine Research Center, School of Public Health, Peking University, No. 38 Xue-Yuan Road, Haidian District, Beijing, 100191, China.
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| |
Collapse
|
21
|
Tomalka J, Sharma A, Smith AGC, Avaliani T, Gujabidze M, Bakuradze T, Sabanadze S, Jones DP, Avaliani Z, Kipiani M, Kempker RR, Collins JM. Combined cerebrospinal fluid metabolomic and cytokine profiling in tuberculosis meningitis reveals robust and prolonged changes in immunometabolic networks. Tuberculosis (Edinb) 2024; 144:102462. [PMID: 38070353 PMCID: PMC10842779 DOI: 10.1016/j.tube.2023.102462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/19/2023]
Abstract
Much of the high mortality in tuberculosis meningitis (TBM) is attributable to excessive inflammation, making it imperative to identify targets for host-directed therapies that reduce pathologic inflammation and mortality. In this study, we investigate how cytokines and metabolites in the cerebral spinal fluid (CSF) associate with TBM at diagnosis and during TBM treatment. At diagnosis, TBM patients (n = 17) demonstrate significant increases of cytokines and chemokines that promote inflammation and cell migration including IL-17A, IL-2, TNFα, IFNγ, and IL-1β versus asymptomatic controls without known central nervous system pathology (n = 20). Inflammatory immune signaling had a strong positive correlation with immunomodulatory metabolites including kynurenine, lactic acid, and carnitine and strong negative correlations with tryptophan and itaconate. Inflammatory immunometabolic networks were only partially reversed with two months of effective TBM treatment and remained significantly different compared to CSF from controls. Together, these data highlight a critical role for host metabolism in regulating the inflammatory response to TBM and indicate the timeline for restoration of immune homeostasis in the CSF is prolonged.
Collapse
Affiliation(s)
- Jeffrey Tomalka
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Ashish Sharma
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Alison G C Smith
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Teona Avaliani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Mariam Gujabidze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Tinatin Bakuradze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Shorena Sabanadze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Dean P Jones
- Division of Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Zaza Avaliani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia; European University, Tbilisi, Georgia
| | - Maia Kipiani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia; The University of Georgia, Tbilisi, Georgia; David Tvildiani Medical University, The University of Georgia, Tbilisi, Georgia
| | - Russell R Kempker
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeffrey M Collins
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
22
|
Collins JM, Kipiani M, Jin Y, Sharma AA, Tomalka JA, Avaliani T, Gujabidze M, Bakuradze T, Sabanadze S, Avaliani Z, Blumberg HM, Benkeser D, Jones DP, Peloquin C, Kempker RR. Pharmacometabolomics in TB Meningitis - understanding the pharmacokinetic, metabolic, and immune factors associated with anti-TB drug concentrations in cerebrospinal fluid. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.14.23299982. [PMID: 38168338 PMCID: PMC10760251 DOI: 10.1101/2023.12.14.23299982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Poor penetration of many anti-tuberculosis (TB) antibiotics into the central nervous system (CNS) is thought to be a major driver of morbidity and mortality in TB meningitis (TBM). While the amount of a particular drug that crosses into the cerebrospinal fluid (CSF) varies from person to person, little is known about the host factors associated with interindividual differences in CSF concentrations of anti-TB drugs. In patients diagnosed with TBM from the country of Georgia (n=17), we investigate the association between CSF concentrations of anti-TB antibiotics and multiple host factors including serum drug concentrations and CSF concentrations of metabolites and cytokines. We found >2-fold differences in CSF concentrations of anti-TB antibiotics from person to person for all drugs tested including cycloserine, ethambutol, imipenem, isoniazid, levofloxacin, linezolid, moxifloxacin pyrazinamide, and rifampin. While serum drug concentrations explained over 40% of the variation in CSF drug concentrations for cycloserine, isoniazid, linezolid, and pyrazinamide (adjusted R 2 >0.4, p<0.001 for all), there was no evidence of an association between serum concentrations of imipenem and ethambutol and their respective CSF concentrations. CSF concentrations of carnitines were significantly associated with concentrations of ethambutol and imipenem (q<0.05), and imipenem was the only antibiotic significantly associated with CSF cytokine concentrations. These results indicate that there is high interindividual variability in CSF drug concentrations in patients treated for TBM, which is only partially explained by differences in serum drug concentrations and not associated with concentrations of cytokines and chemokines in the CSF.
Collapse
|
23
|
Konas D, Cho S, Thomas OD, Bhatti MM, Leon Hernandez K, Moran C, Booter H, Candela T, Lacap J, McFadden P, van den Berg S, Welter AM, Peralta A, Janson CA, Catalano J, Goodey NM. Investigating the Roles of Active Site Residues in Mycobacterium tuberculosis Indole-3-glycerol Phosphate Synthase, a Potential Target for Antitubercular Agents. ACS BIO & MED CHEM AU 2023; 3:438-447. [PMID: 37876495 PMCID: PMC10591298 DOI: 10.1021/acsbiomedchemau.3c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 10/26/2023]
Abstract
Mycobacterium tuberculosis drug resistance is emerging and new drug targets are needed. Tryptophan biosynthesis is necessary for M. tuberculosis replication and virulence. Indole-3-glycerol phosphate synthase (IGPS) catalyzes a step in M. tuberculosis tryptophan biosynthesis and has been suggested as a potential anti-infective target, but our understanding of this enzyme is limited. To aid in inhibitor design and gain a greater mechanistic picture of this enzyme, there is a need to understand the roles of active site amino acids in ligand binding and catalysis. In this work, we explored the roles of conserved active site amino acids Glu57, Lys59, Lys119, Glu168, and Glu219. Mutation of each to Ala results in loss of all detectable activity. The Glu57Gln, Lys59Arg, Lys119Arg, Glu168Gln, and Glu219Asp mutations result in large activity losses, while Glu219Gln has enhanced activity. Analysis of the enzymatic data yields the following main conclusions: (A) Lys119 is the likely catalytic acid in the CdRP ring closure step. (B) Glu168 stabilizes a charged reaction intermediate and may also be the catalytic base. (C) Glu57, Glu219, and Lys119 form a closely arranged triad in which Glu57 and Glu219 modulate the pKa of Lys119, and thus overall activity. This increased understanding of inter- and intramolecular interactions and demonstration of the highly coordinated nature of the M. tuberculosis IGPS active site provide new mechanistic information and guidance for future work with this potential new drug target.
Collapse
Affiliation(s)
- David
W. Konas
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Sarah Cho
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Oshane D. Thomas
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Maryum M. Bhatti
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Katherine Leon Hernandez
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Cinthya Moran
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Hedda Booter
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Thomas Candela
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Joseph Lacap
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Paige McFadden
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Savannah van den Berg
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Alyssa M. Welter
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Ashley Peralta
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Cheryl A. Janson
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Jaclyn Catalano
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| | - Nina M. Goodey
- Department of Chemistry and
Biochemistry, Montclair State University 1 Normal Avenue, Montclair, New Jersey 07043, United States
| |
Collapse
|
24
|
Kaushal D, Singh DK, Mehra S. Immune Responses in Lung Granulomas during Mtb/HIV Co-Infection: Implications for Pathogenesis and Therapy. Pathogens 2023; 12:1120. [PMID: 37764928 PMCID: PMC10534770 DOI: 10.3390/pathogens12091120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
HIV and TB are the cause of significant worldwide mortality and pose a grave danger to the global public health. TB is the leading cause of death in HIV-infected persons, with one in four deaths attributable to TB. While the majority of healthy individuals infected with M. tuberculosis (Mtb) are able to control the infection, co-infection with HIV increases the risk of TB infection progressing to TB disease by over 20-fold. While antiretroviral therapy (ART), the cornerstone of HIV care, decreases the incidence of TB in HIV-uninfected people, this remains 4- to 7-fold higher after ART in HIV-co-infected individuals in TB-endemic settings, regardless of the duration of therapy. Thus, the immune control of Mtb infection in Mtb/HIV-co-infected individuals is not fully restored by ART. We do not fully understand the reasons why Mtb/HIV-co-infected individuals maintain a high susceptibility to the reactivation of LTBI, despite an effective viral control by ART. A deep understanding of the molecular mechanisms that govern HIV-induced reactivation of TB is essential to develop improved treatments and vaccines for the Mtb/HIV-co-infected population. We discuss potential strategies for the mitigation of the observed chronic immune activation in combination with both anti-TB and anti-retroviral approaches.
Collapse
Affiliation(s)
| | | | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| |
Collapse
|
25
|
Herbert C, Luies L, Loots DT, Williams AA. The metabolic consequences of HIV/TB co-infection. BMC Infect Dis 2023; 23:536. [PMID: 37592227 PMCID: PMC10436461 DOI: 10.1186/s12879-023-08505-4] [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: 04/26/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND The synergy between the human immunodeficiency virus (HIV) and Mycobacterium tuberculosis during co-infection of a host is well known. While this synergy is known to be driven by immunological deterioration, the metabolic mechanisms that contribute to the associated disease burden experienced during HIV/tuberculosis (TB) co-infection remain poorly understood. Furthermore, while anti-HIV treatments suppress viral replication, these therapeutics give rise to host metabolic disruption and adaptations beyond that induced by only infection or disease. METHODS In this study, the serum metabolic profiles of healthy controls, untreated HIV-negative TB-positive patients, untreated HIV/TB co-infected patients, and HIV/TB co-infected patients on antiretroviral therapy (ART), were measured using two-dimensional gas chromatography time-of-flight mass spectrometry. Since no global metabolic profile for HIV/TB co-infection and the effect of ART has been published to date, this pilot study aimed to elucidate the general areas of metabolism affected during such conditions. RESULTS HIV/TB co-infection induced significant changes to the host's lipid and protein metabolism, with additional microbial product translocation from the gut to the blood. The results suggest that HIV augments TB synergistically, at least in part, contributing to increased inflammation, oxidative stress, ART-induced mitochondrial damage, and its detrimental effects on gut health, which in turn, affects energy availability. ART reverses these trends to some extent in HIV/TB co-infected patients but not to that of healthy controls. CONCLUSION This study generated several new hypotheses that could direct future metabolic studies, which could be combined with other research techniques or methodologies to further elucidate the underlying mechanisms of these changes.
Collapse
Affiliation(s)
- Chandré Herbert
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Laneke Luies
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | | |
Collapse
|
26
|
Tomalka J, Sharma A, Smith AGC, Avaliani T, Gujabidze M, Bakuradze T, Sabanadze S, Jones DP, Avaliani Z, Kipiani M, Kempker RR, Collins JM. Combined cerebrospinal fluid metabolomic and cytokine profiling in tuberculosis meningitis reveals robust and prolonged changes in immunometabolic networks. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.26.23291676. [PMID: 37425849 PMCID: PMC10327257 DOI: 10.1101/2023.06.26.23291676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Much of the high mortality in tuberculosis meningitis (TBM) is attributable to excessive inflammation, making it imperative to identify targets for host-directed therapies that reduce pathologic inflammation and mortality. In this study, we investigate how cytokines and metabolites in the cerebral spinal fluid (CSF) associate with TBM at diagnosis and during TBM treatment. At diagnosis, TBM patients demonstrate significant increases versus controls of cytokines and chemokines that promote inflammation and cell migration including IL-17A, IL-2, TNFα, IFNγ, and IL-1β. Inflammatory immune signaling was strongly correlated with immunomodulatory metabolites including kynurenine, lactic acid, carnitine, tryptophan, and itaconate. Inflammatory immunometabolic networks were only partially reversed with two months of effective TBM treatment and remained significantly different versus control CSF. Together, these data highlight a critical role for host metabolism in regulating the inflammatory response to TBM and indicate the timeline for restoration of immune homeostasis in the CSF is prolonged.
Collapse
|
27
|
Beukes D, van Reenen M, Loots DT, du Preez I. Tuberculosis is associated with sputum metabolome variations, irrespective of patient sex or HIV status: an untargeted GCxGC-TOFMS study. Metabolomics 2023; 19:55. [PMID: 37284915 DOI: 10.1007/s11306-023-02017-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
INTRODUCTION Various studies have identified TB-induced metabolome variations. However, in most of these studies, a large degree of variation exists between individual patients. OBJECTIVES To identify differential metabolites for TB, independent of patients' sex or HIV status. METHODS Untargeted GCxGC/TOF-MS analyses were applied to the sputum of 31 TB + and 197 TB- individuals. Univariate statistics were used to identify metabolites which are significantly different between TB + and TB- individuals (a) irrespective of HIV status, and (b) with a HIV + status. Comparisons a and b were repeated for (i) all participants, (ii) males only and (iii) females only. RESULTS Twenty-one compounds were significantly different between the TB + and TB- individuals within the female subgroup (11% lipids; 10% carbohydrates; 1% amino acids, 5% other and 73% unannotated), and 6 within the male subgroup (20% lipids; 40% carbohydrates; 6% amino acids, 7% other and 27% unannotated). For the HIV + patients (TB + vs. TB-), a total of 125 compounds were significant within the female subgroup (16% lipids; 8% carbohydrates; 12% amino acids, 6% organic acids, 8% other and 50% unannotated), and 44 within the male subgroup (17% lipids; 2% carbohydrates; 14% amino acids related, 8% organic acids, 9% other and 50% unannotated). Only one annotated compound, 1-oleoyl lysophosphaditic acid, was consistently identified as a differential metabolite for TB, irrespective of sex or HIV status. The potential clinical application of this compound should be evaluated further. CONCLUSIONS Our findings highlight the importance of considering confounders in metabolomics studies in order to identify unambiguous disease biomarkers.
Collapse
Affiliation(s)
- Derylize Beukes
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Mari van Reenen
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Ilse du Preez
- Human Metabolomics, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
28
|
Wang H, de Carvalho LPS. Metabolomic profiling reveals bacterial metabolic adaptation strategies and new metabolites. Curr Opin Chem Biol 2023; 74:102287. [PMID: 36948086 DOI: 10.1016/j.cbpa.2023.102287] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/07/2023] [Accepted: 02/21/2023] [Indexed: 03/24/2023]
Abstract
How has metabolomics helped our understanding of infectious diseases? With the threat of antimicrobial resistance to human health around the world, metabolomics has emerged as a powerful tool to comprehensively characterize metabolic pathways to identify new drug targets. However, its output is constrained to known metabolites and their metabolic pathways. Recent advances in instrumentation, methodologies, and computational mass spectrometry have accelerated the use of metabolomics to understand pathogen-host metabolic interactions. This short review discusses a selection of recent publications using metabolomics in infectious/bacterial diseases. These studies unravel the links between metabolic adaptations to environments and host metabolic responses. Moreover, they highlight the importance of enzyme function and metabolite characterization in identifying new drug targets and biomarkers, as well as precision medicine in monitoring therapeutics and diagnosing diseases.
Collapse
Affiliation(s)
- Hua Wang
- Pigments of Life Research Laboratory, School of Infection & Immunity, University of Glasgow, Glasgow G12 8TA, United Kingdom.
| | - Luiz Pedro S de Carvalho
- Mycobacterial Metabolism and Antibiotic Research Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter 33458, United States.
| |
Collapse
|
29
|
Singh B, Moodley C, Singh DK, Escobedo RA, Sharan R, Arora G, Ganatra SR, Shivanna V, Gonzalez O, Hall-Ursone S, Dick EJ, Kaushal D, Alvarez X, Mehra S. Inhibition of indoleamine dioxygenase leads to better control of tuberculosis adjunctive to chemotherapy. JCI Insight 2023; 8:e163101. [PMID: 36692017 PMCID: PMC9977315 DOI: 10.1172/jci.insight.163101] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/30/2022] [Indexed: 01/24/2023] Open
Abstract
The expression of indoleamine 2,3-dioxygenase (IDO), a robust immunosuppressant, is significantly induced in macaque tuberculosis (TB) granulomas, where it is expressed on IFN-responsive macrophages and myeloid-derived suppressor cells. IDO expression is also highly induced in human TB granulomas, and products of its activity are detected in patients with TB. In vivo blockade of IDO activity resulted in the reorganization of the granuloma with substantially greater T cells being recruited to the core of the lesions. This correlated with better immune control of TB and reduced lung M. tuberculosis burdens. To study if the IDO blockade strategy can be translated to a bona fide host-directed therapy in the clinical setting of TB, we studied the effect of IDO inhibitor 1-methyl-d-tryptophan adjunctive to suboptimal anti-TB chemotherapy. While two-thirds of controls and one-third of chemotherapy-treated animals progressed to active TB, inhibition of IDO adjunctive to the same therapy protected macaques from TB, as measured by clinical, radiological, and microbiological attributes. Although chemotherapy improved proliferative T cell responses, adjunctive inhibition of IDO further enhanced the recruitment of effector T cells to the lung. These results strongly suggest the possibility that IDO inhibition can be attempted adjunctive to anti-TB chemotherapy in clinical trials.
Collapse
|
30
|
Kumar NP, Nancy A, Viswanathan V, Sivakumar S, Thiruvengadam K, Ahamed SF, Hissar S, Kornfeld H, Babu S. Chitinase and indoleamine 2, 3-dioxygenase are prognostic biomarkers for unfavorable treatment outcomes in pulmonary tuberculosis. Front Immunol 2023; 14:1093640. [PMID: 36814914 PMCID: PMC9939892 DOI: 10.3389/fimmu.2023.1093640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/17/2023] [Indexed: 02/08/2023] Open
Abstract
Introduction Chitinase, Indoleamine 2,3-dioxygenesae-1 (IDO-1) and heme oxygenase-1 (HO-1) are candidate diagnostic biomarkers for tuberculosis (TB). Whether these immune markers could also serve as predictive biomarkers of unfavorable treatment outcomes in pulmonary TB (PTB) is not known. Methods A cohort of newly diagnosed, sputum culture-positive adults with drug-sensitive PTB were recruited. Plasma chitinase protein, IDO protein and HO-1 levels measured before treatment initiation were compared between 68 cases with unfavorable outcomes (treatment failure, death, or recurrence) and 108 control individuals who had recurrence-free cure. Results Plasma chitinase and IDO protein levels but not HO-1 levels were lower in cases compared to controls. The low chitinase and IDO protein levels were associated with increased risk of unfavourable outcomes in unadjusted and adjusted analyses. Receiver operating characteristic analysis revealed that chitinase and IDO proteins exhibited high sensitivity and specificity in differentiating cases vs controls as well as in differentiating treatment failure vs controls and recurrence vs controls, respectively. Classification and regression trees (CART) were used to determine threshold values for these two immune markers. Discussion Our study revealed a plasma chitinase and IDO protein signature that may be used as a tool for predicting adverse treatment outcomes in PTB.
Collapse
Affiliation(s)
- Nathella Pavan Kumar
- Department of Immunology, National Institute for Research in Tuberculosis, Indian Council of Medical Research (ICMR), Chennai, India
| | - Arul Nancy
- International Center for Excellence in Research, National Institutes of Health, National Institute for Research in Tuberculosis (NIRT), International Center for Excellence in Research, Chennai, India
| | - Vijay Viswanathan
- Diabetology, Prof. M. Viswanathan Diabetes Research Center, Chennai, India
| | - Shanmugam Sivakumar
- Department of Bacteriology, National Institute for Research in Tuberculosis, Indian Council of Medical Research (ICMR), Chennai, India
| | - Kannan Thiruvengadam
- Epidemiology Statistics, National Institute for Research in Tuberculosis, Indian Council of Medical Research (ICMR), Chennai, India
| | - Shaik Fayaz Ahamed
- International Center for Excellence in Research, National Institutes of Health, National Institute for Research in Tuberculosis (NIRT), International Center for Excellence in Research, Chennai, India
| | - Syed Hissar
- Clinical Research, National Institute for Research in Tuberculosis, Indian Council of Medical Research (ICMR), Chennai, India
| | - Hardy Kornfeld
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Subash Babu
- International Center for Excellence in Research, National Institutes of Health, National Institute for Research in Tuberculosis (NIRT), International Center for Excellence in Research, Chennai, India.,Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
31
|
Yu Y, Jiang XX, Li JC. Biomarker discovery for tuberculosis using metabolomics. Front Mol Biosci 2023; 10:1099654. [PMID: 36891238 PMCID: PMC9986447 DOI: 10.3389/fmolb.2023.1099654] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Tuberculosis (TB) is the leading cause of death among infectious diseases, and the ratio of cases in which its pathogen Mycobacterium tuberculosis (Mtb) is drug resistant has been increasing worldwide, whereas latent tuberculosis infection (LTBI) may develop into active TB. Thus it is important to understand the mechanism of drug resistance, find new drugs, and find biomarkers for TB diagnosis. The rapid progress of metabolomics has enabled quantitative metabolite profiling of both the host and the pathogen. In this context, we provide recent progress in the application of metabolomics toward biomarker discovery for tuberculosis. In particular, we first focus on biomarkers based on blood or other body fluids for diagnosing active TB, identifying LTBI and predicting the risk of developing active TB, as well as monitoring the effectiveness of anti-TB drugs. Then we discuss the pathogen-based biomarker research for identifying drug resistant TB. While there have been many reports of potential candidate biomarkers, validations and clinical testing as well as improved bioinformatics analysis are needed to further substantiate and select key biomarkers before they can be made clinically applicable.
Collapse
Affiliation(s)
- Yi Yu
- Center for Analyses and Measurements, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Xin-Xin Jiang
- Clinical Research Laboratory, Shaoxing Seventh People's Hospital, Shaoxing, China
| | - Ji-Cheng Li
- Clinical Research Laboratory, Shaoxing Seventh People's Hospital, Shaoxing, China.,Institute of Cell Biology, Zhejiang University Medical School, Hangzhou, China
| |
Collapse
|
32
|
Rawat BS, Kumar D, Soni V, Rosenn EH. Therapeutic Potentials of Immunometabolomic Modulations Induced by Tuberculosis Vaccination. Vaccines (Basel) 2022; 10:vaccines10122127. [PMID: 36560537 PMCID: PMC9781011 DOI: 10.3390/vaccines10122127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Metabolomics is emerging as a promising tool to understand the effect of immunometabolism for the development of novel host-directed alternative therapies. Immunometabolism can modulate both innate and adaptive immunity in response to pathogens and vaccinations. For instance, infections can affect lipid and amino acid metabolism while vaccines can trigger bile acid and carbohydrate pathways. Metabolomics as a vaccinomics tool, can provide a broader picture of vaccine-induced biochemical changes and pave a path to potentiate the vaccine efficacy. Its integration with other systems biology tools or treatment modes can enhance the cure, response rate, and control over the emergence of drug-resistant strains. Mycobacterium tuberculosis (Mtb) infection can remodel the host metabolism for its survival, while there are many biochemical pathways that the host adjusts to combat the infection. Similarly, the anti-TB vaccine, Bacillus Calmette-Guerin (BCG), was also found to affect the host metabolic pathways thus modulating immune responses. In this review, we highlight the metabolomic schema of the anti-TB vaccine and its therapeutic applications. Rewiring of immune metabolism upon BCG vaccination induces different signaling pathways which lead to epigenetic modifications underlying trained immunity. Metabolic pathways such as glycolysis, central carbon metabolism, and cholesterol synthesis play an important role in these aspects of immunity. Trained immunity and its applications are increasing day by day and it can be used to develop the next generation of vaccines to treat various other infections and orphan diseases. Our goal is to provide fresh insight into this direction and connect various dots to develop a conceptual framework.
Collapse
Affiliation(s)
- Bhupendra Singh Rawat
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Deepak Kumar
- Department of Zoology, University of Rajasthan, Jaipur 302004, Rajasthan, India
| | - Vijay Soni
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Correspondence:
| | - Eric H. Rosenn
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
33
|
Zhang Y, Yue Q, Zhu H, Song J, Li D, Liu W, Jiang S, Jiang N, Qiu C, Ai J, Zhang Y, Zhang W. Serum Metabolic Correlates of the Antibody Response in Subjects Receiving the Inactivated COVID-19 Vaccine. Vaccines (Basel) 2022; 10:1890. [PMID: 36366398 PMCID: PMC9699138 DOI: 10.3390/vaccines10111890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Metabolites are involved in biological process that govern the immune response to infection and vaccination. Knowledge of how metabolites interact with the immune system during immunization with the COVID-19 vaccine is limited. Here, we report that the serum metabolites are correlated with the magnitude of the antibody response in recipients receiving the inactivated COVID-19 vaccine, which provides critical information for studying metabolism regarding the human immune response to vaccination. METHODS 106 healthy volunteers without history of SARS-CoV-2 infection or vaccination were prospectively enrolled to receive the primary series of two doses of inactivated whole-virion SARS-CoV-2 vaccine. The serum samples were collected 2-4 weeks after the second dose. The magnitude of the anti-RBD antibody was quantified using surrogate virus neutralization tests. The profile of metabolites in serum was identified using untargeted metabolomics analysis. RESULTS The level of anti-RBD antibody 14-28 days after the second dose was significantly elevated and its interpersonal variability was diverse in a wide range. Thirty-two samples at extremes of the anti-RBD antibody titer were selected to discover the metabolic correlates. Two hundred and fifteen differential metabolites associated with antibody response independent of body mass index were identified. Pregnenolone and sphingolipid metabolism might be involved in the modulation of the human antibody response to the inactivated COVID-19 vaccine. CONCLUSION We discovered key metabolites as well as those with a related functional significance that might modulate the human immune response to vaccination.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qiaoyan Yue
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210006, China
| | - Haojing Zhu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210006, China
| | - Jieyu Song
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Dingding Li
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210006, China
| | - Wen Liu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210006, China
| | - Shujun Jiang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210006, China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210006, China
| | - Ning Jiang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chao Qiu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jingwen Ai
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yanliang Zhang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210006, China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210006, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Huashen Institute of Microbes and Infections, No. 6 Lane 1220 Huashan Rd., Shanghai 200031, China
| |
Collapse
|
34
|
Kim J, Suresh B, Lim MN, Hong SH, Kim KS, Song HE, Lee HY, Yoo HJ, Kim WJ. Metabolomics Reveals Dysregulated Sphingolipid and Amino Acid Metabolism Associated with Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2022; 17:2343-2353. [PMID: 36172036 PMCID: PMC9511892 DOI: 10.2147/copd.s376714] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease presenting as multiple phenotypes, such as declining lung function, emphysema, or persistent airflow limitation caused by several risk factors, including cigarette smoking and air pollution. The inherent complexity of COPD phenotypes propounds difficulties for accurate diagnosis and prognosis. Although metabolomic profiles on COPD have been reported, the role of metabolism in COPD-related phenotypes is yet to be determined. In this study, we investigated the association between plasma sphingolipids and amino acids, and between COPD and COPD-related phenotypes in a Korean cohort. Patients and Methods Blood samples were collected from 120 patients with COPD and 80 control participants who underwent spirometry and quantitative computed tomography. The plasma metabolic profiling was carried out using LC-MS/MS analysis. Results Among the evaluated plasma sphingolipids, an increase in the metabolism of two specific sphingomyelins, SM (d18:1/24:0) and SM (d18:1/24:1) were significantly associated with COPD. There was no significant correlation between any of the SMs and the emphysema index, FVC and FEV1 in the COPD cohort. Meanwhile, Cer (d18:1/18:0) and Cer (d18:1/24:1) were significantly associated with reduced FEV1. Furthermore, the levels of several amino acids were altered in the COPD group compared to that in the non-COPD group; glutamate and alpha AAA were substantial associated with emphysema in COPD. Kynurenine was the only amino acid significantly associated with reduced FEV1 in COPD. In contrast, there was no correlation between FVC and the elevated metabolites. Conclusion Our results provide dysregulated plasma metabolites impacting COPD phenotypes, although more studies are needed to explore the underlying mechanism related to COPD pathogenesis.
Collapse
Affiliation(s)
- Jeeyoung Kim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Bharathi Suresh
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Myoung Nam Lim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea.,College of Medicine, Hanyang University, Seoul, South Korea
| | - Ha Eun Song
- Department of Convergence Medicine, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyo Yeong Lee
- Department of Convergence Medicine, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Woo Jin Kim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
35
|
Amalia F, Syamsunarno MRAA, Triatin RD, Fatimah SN, Chaidir L, Achmad TH. The Role of Amino Acids in Tuberculosis Infection: A Literature Review. Metabolites 2022; 12:933. [PMID: 36295834 PMCID: PMC9611225 DOI: 10.3390/metabo12100933] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022] Open
Abstract
Recently, there was an abundance of studies being conducted on the metabolomic profiling of tuberculosis patients. Amino acids are critical metabolites for the immune system, as they might contribute to providing nutrients for the host intracellular pathway. In tuberculosis, several amino acids play important roles in both the mycobacteria infection mechanism and the host. Individual studies showed how the dynamics of metabolite products that result from interactions between Mycobacterium tuberculosis (Mtb) and the host play important roles in different stages of infection. In this review, we focus on the dynamics of amino-acid metabolism and identify the prominent roles of amino acids in the diagnostics and treatment of tuberculosis infection. Online resources, including PubMed, ScienceDirect, Scopus, and Clinical Key, were used to search for articles with combination keywords of amino acids and TB. The inclusion criteria were full-text articles in English published in the last 10 years. Most amino acids were decreased in patients with active TB compared with those with latent TB and healthy controls. However, some amino acids, including leucine, isoleucine, valine, phenylalanine, aspartate, and glutamate, were found to be at higher levels in TB patients. Additionally, the biomarkers of Mtb infection included the ratios of kynurenine to tryptophan, phenylalanine to histidine, and citrulline to arginine. Most amino acids were present at different levels in different stages of infection and disease progression. The search for additional roles played by those metabolomic biomarkers in each stage of infection might facilitate diagnostic tools for staging TB infection.
Collapse
Affiliation(s)
- Fiki Amalia
- Study Program of Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung 40161, Jawa Barat, Indonesia
| | - Mas Rizky A. A. Syamsunarno
- Department of Biomedical Sciences, Faculty of Medicine Universitas Padjadjaran, Bandung 40161, Jawa Barat, Indonesia
- Center for Translational Biomarker Research, Universitas Padjadjaran, Bandung 40161, Jawa Barat, Indonesia
| | - Rima Destya Triatin
- Department of Biomedical Sciences, Faculty of Medicine Universitas Padjadjaran, Bandung 40161, Jawa Barat, Indonesia
| | - Siti Nur Fatimah
- Department of Public Health, Faculty of Medicine Universitas Padjadjaran, Bandung 40161, Jawa Barat, Indonesia
| | - Lidya Chaidir
- Department of Biomedical Sciences, Faculty of Medicine Universitas Padjadjaran, Bandung 40161, Jawa Barat, Indonesia
- Center for Translational Biomarker Research, Universitas Padjadjaran, Bandung 40161, Jawa Barat, Indonesia
| | - Tri Hanggono Achmad
- Department of Biomedical Sciences, Faculty of Medicine Universitas Padjadjaran, Bandung 40161, Jawa Barat, Indonesia
| |
Collapse
|
36
|
Heyckendorf J, Georghiou SB, Frahm N, Heinrich N, Kontsevaya I, Reimann M, Holtzman D, Imperial M, Cirillo DM, Gillespie SH, Ruhwald M, on behalf of the UNITE4TB Consortium. Tuberculosis Treatment Monitoring and Outcome Measures: New Interest and New Strategies. Clin Microbiol Rev 2022; 35:e0022721. [PMID: 35311552 PMCID: PMC9491169 DOI: 10.1128/cmr.00227-21] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Despite the advent of new diagnostics, drugs and regimens, tuberculosis (TB) remains a global public health threat. A significant challenge for TB control efforts has been the monitoring of TB therapy and determination of TB treatment success. Current recommendations for TB treatment monitoring rely on sputum and culture conversion, which have low sensitivity and long turnaround times, present biohazard risk, and are prone to contamination, undermining their usefulness as clinical treatment monitoring tools and for drug development. We review the pipeline of molecular technologies and assays that serve as suitable substitutes for current culture-based readouts for treatment response and outcome with the potential to change TB therapy monitoring and accelerate drug development.
Collapse
Affiliation(s)
- Jan Heyckendorf
- Department of Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
| | | | - Nicole Frahm
- Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, USA
| | - Norbert Heinrich
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
| | - Irina Kontsevaya
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
| | - Maja Reimann
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
- International Health/Infectious Diseases, University of Lübeck, Lübeck, Germany
| | - David Holtzman
- FIND, the Global Alliance for Diagnostics, Geneva, Switzerland
| | - Marjorie Imperial
- University of California San Francisco, San Francisco, California, USA, United States
| | - Daniela M. Cirillo
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stephen H. Gillespie
- School of Medicine, University of St Andrewsgrid.11914.3c, St Andrews, Fife, Scotland
| | - Morten Ruhwald
- FIND, the Global Alliance for Diagnostics, Geneva, Switzerland
| | | |
Collapse
|
37
|
Gough M, Singh DK, Moodley C, Niu T, Golden NA, Kaushal D, Mehra S. Peripheral Blood Markers Correlate with the Progression of Active Tuberculosis Relative to Latent Control of Mycobacterium tuberculosis Infection in Macaques. Pathogens 2022; 11:544. [PMID: 35631065 PMCID: PMC9146669 DOI: 10.3390/pathogens11050544] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 01/08/2023] Open
Abstract
Despite a century of research into tuberculosis (TB), there is a dearth of reproducible, easily quantifiable, biomarkers that can predict disease onset and differentiate between host disease states. Due to the challenges associated with human sampling, nonhuman primates (NHPs) are utilized for recapitulating the closest possible modelling of human TB. To establish a predictive peripheral biomarker profile based on a larger cohort of rhesus macaques (RM), we analyzed results pertaining to peripheral blood serum chemistry and cell counts from RMs that were experimentally exposed to Mtb in our prior studies and characterized as having either developed active TB (ATB) disease or latent TB infection (LTBI). We compared lung CFU burdens and quantitative pathologies with a number of measurables in the peripheral blood. Based on our results, the investigations were then extended to the study of specific molecules and cells in the lung compartments of a subset of these animals and their immune responses. In addition to the elevated serum C-reactive protein (CRP) levels, frequently used to discern the level of Mtb infection in model systems, reduced serum albumin-to-globulin (A/G) ratios were also predictive of active TB disease. Furthermore, higher peripheral myeloid cell levels, particularly those of neutrophils, kynurenine-to-tryptophan ratio, an indicator of induced expression of the immunosuppressive molecule indoleamine dioxygenase, and an influx of myeloid cell populations could also efficiently discriminate between ATB and LTBI in experimentally infected macaques. These quantifiable correlates of disease were then used in conjunction with a regression-based analysis to predict bacterial load. Our results suggest a potential biomarker profile of TB disease in rhesus macaques, that could inform future NHP-TB research. Our results thus suggest that specific biomarkers may be developed from the myeloid subset of peripheral blood or plasma with the ability to discriminate between active and latent Mtb infection.
Collapse
Affiliation(s)
- Maya Gough
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
| | - Dhiraj K. Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
| | - Chivonne Moodley
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
- Tulane University Health Science Center, New Orleans, LA 70112, USA
| | - Tianhua Niu
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Nadia A. Golden
- Tulane National Primate Research Center, Covington, LA 70433, USA;
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
| |
Collapse
|
38
|
Tornheim JA, Paradkar M, Zhao H, Kulkarni V, Pradhan N, Kinikar A, Kagal A, Gupte N, Mave V, Gupta A, Karakousis PC. The Kynurenine/Tryptophan Ratio Is a Sensitive Biomarker for the Diagnosis of Pediatric Tuberculosis Among Indian Children. Front Immunol 2022; 12:774043. [PMID: 35095848 PMCID: PMC8790563 DOI: 10.3389/fimmu.2021.774043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/14/2021] [Indexed: 11/30/2022] Open
Abstract
Objectives Pediatric tuberculosis (TB) remains difficult to diagnose. The plasma kynurenine to tryptophan ratio (K/T ratio) is a potential biomarker for TB diagnosis and treatment response but has not been assessed in children. Methods We performed a targeted diagnostic accuracy analysis of four biomarkers: kynurenine abundance, tryptophan abundance, the K/T ratio, and IDO-1 gene expression. Data were obtained from transcriptome and metabolome profiling of children with confirmed tuberculosis and age- and sex-matched uninfected household contacts of pulmonary tuberculosis patients. Each biomarker was assessed as a baseline diagnostic and in response to successful TB treatment. Results Despite non-significant between-group differences in unbiased analysis, the K/T ratio achieved an area under the receiver operator characteristic curve (AUC) of 0.667 and 81.5% sensitivity for TB diagnosis. Kynurenine, tryptophan, and IDO-1 demonstrated diagnostic AUCs of 0.667, 0.602, and 0.463, respectively. None of these biomarkers demonstrated high AUCs for treatment response. The AUC of the K/T ratio was lower than biomarkers identified in unbiased analysis, but improved sensitivity over existing commercial assays for pediatric TB diagnosis. Conclusions Plasma kynurenine and the K/T ratio may be useful biomarkers for pediatric TB. Ongoing studies in geographically diverse populations will determine optimal use of these biomarkers worldwide.
Collapse
Affiliation(s)
- Jeffrey A Tornheim
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Center for Clinical Global Health Education, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mandar Paradkar
- Byramjee Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Henry Zhao
- Johns Hopkins University, Baltimore, MD, United States
| | - Vandana Kulkarni
- Byramjee Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Neeta Pradhan
- Byramjee Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Aarti Kinikar
- Byramjee Jeejeebhoy Government Medical College, Pune, India
| | - Anju Kagal
- Byramjee Jeejeebhoy Government Medical College, Pune, India
| | - Nikhil Gupte
- Center for Clinical Global Health Education, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Byramjee Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Vidya Mave
- Center for Clinical Global Health Education, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Byramjee Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Amita Gupta
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Center for Clinical Global Health Education, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Petros C Karakousis
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Center for Clinical Global Health Education, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
39
|
Abstract
Tuberculosis (TB) in humans is characterized by formation of immune-rich granulomas in infected tissues, the architecture and composition of which are thought to affect disease outcome. However, our understanding of the spatial relationships that control human granulomas is limited. Here, we used multiplexed ion beam imaging by time of flight (MIBI-TOF) to image 37 proteins in tissues from patients with active TB. We constructed a comprehensive atlas that maps 19 cell subsets across 8 spatial microenvironments. This atlas shows an IFN-γ-depleted microenvironment enriched for TGF-β, regulatory T cells and IDO1+ PD-L1+ myeloid cells. In a further transcriptomic meta-analysis of peripheral blood from patients with TB, immunoregulatory trends mirror those identified by granuloma imaging. Notably, PD-L1 expression is associated with progression to active TB and treatment response. These data indicate that in TB granulomas, there are local spatially coordinated immunoregulatory programs with systemic manifestations that define active TB.
Collapse
|
40
|
Shivakoti R, Newman JW, Hanna LE, Queiroz ATL, Borkowski K, Gupte AN, Paradkar M, Satyamurthi P, Kulkarni V, Selva M, Pradhan N, Shivakumar SVBY, Natarajan S, Karunaianantham R, Gupte N, Thiruvengadam K, Fiehn O, Bharadwaj R, Kagal A, Gaikwad S, Sangle S, Golub JE, Andrade BB, Mave V, Gupta A, Padmapriyadarsini C. Host lipidome and tuberculosis treatment failure. Eur Respir J 2022; 59:2004532. [PMID: 34375300 PMCID: PMC9625841 DOI: 10.1183/13993003.04532-2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/24/2021] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Host lipids play important roles in tuberculosis (TB) pathogenesis. Whether host lipids at TB treatment initiation (baseline) affect subsequent treatment outcomes has not been well characterised. We used unbiased lipidomics to study the prospective association of host lipids with TB treatment failure. METHODS A case-control study (n=192), nested within a prospective cohort study, was used to investigate the association of baseline plasma lipids with TB treatment failure among adults with pulmonary TB. Cases (n=46) were defined as TB treatment failure, while controls (n=146) were those without failure. Complex lipids and inflammatory lipid mediators were measured using liquid chromatography mass spectrometry techniques. Adjusted least-square regression was used to assess differences in groups. In addition, machine learning identified lipids with highest area under the curve (AUC) to classify cases and controls. RESULTS Baseline levels of 32 lipids differed between controls and those with treatment failure after false discovery rate adjustment. Treatment failure was associated with lower baseline levels of cholesteryl esters and oxylipin, and higher baseline levels of ceramides and triglycerides compared to controls. Two cholesteryl ester lipids combined in a unique classifier model provided an AUC of 0.79 (95% CI 0.65-0.93) in the test dataset for prediction of TB treatment failure. CONCLUSIONS We identified lipids, some with known roles in TB pathogenesis, associated with TB treatment failure. In addition, a lipid signature with prognostic accuracy for TB treatment failure was identified. These lipids could be potential targets for risk-stratification, adjunct therapy and treatment monitoring.
Collapse
Affiliation(s)
- Rupak Shivakoti
- Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Dept of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - John W Newman
- Obesity and Metabolism Research Unit, Western Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Davis, CA, USA
- Dept of Nutrition, University of California, Davis, CA, USA
- West Coast Metabolomics Center, University of California, Davis, CA, USA
| | | | - Artur T L Queiroz
- Instituto Goncalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research, Salvador, Brazil
| | - Kamil Borkowski
- West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Akshay N Gupte
- Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mandar Paradkar
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | | | - Vandana Kulkarni
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Murugesh Selva
- National Institute for Research in Tuberculosis, Chennai, India
| | - Neeta Pradhan
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | | | | | | | - Nikhil Gupte
- Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | | | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Renu Bharadwaj
- Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Anju Kagal
- Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Sanjay Gaikwad
- Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | | | - Jonathan E Golub
- Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bruno B Andrade
- Instituto Goncalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil
- Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Laureate International Universities, Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| | - Vidya Mave
- Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Amita Gupta
- Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
- Equal contribution
| | | |
Collapse
|
41
|
Consalvi S, Venditti G, Zhu J, Boshoff HI, Arora K, De Logu A, Ioerger TR, Rubin EJ, Biava M, Poce G. 6-Fluorophenylbenzohydrazides inhibit Mycobacterium tuberculosis growth through alteration of tryptophan biosynthesis. Eur J Med Chem 2021; 226:113843. [PMID: 34520959 PMCID: PMC10994514 DOI: 10.1016/j.ejmech.2021.113843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/31/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022]
Abstract
A major constraint in reducing tuberculosis epidemic is the emergence of strains resistant to one or more of clinically approved antibiotics, which emphasizes the need of novel drugs with novel targets. Genetic knockout strains of Mycobacterium tuberculosis (Mtb) have established that tryptophan (Trp) biosynthesis is essential for the bacterium to survive in vivo and cause disease in animal models. An anthranilate-like compound, 6-FABA, was previously shown to synergize with the host immune response to Mtb infection in vivo. Herein, we present a class of anthranilate-like compounds endowed with good antimycobacterial activity and low cytotoxicity. We show how replacing the carboxylic moiety with a hydrazide led to a significant improvement in both activity and cytotoxicity relative to the parent compound 6-FABA. Several new benzohydrazides (compounds 20-31, 33, 34, 36, 38 and 39) showed good activities against Mtb (0.625 ≤ MIC≤6.25 μM) and demonstrated no detectable cytotoxicity against Vero cell assay (CC50 ≥ 1360 μM). The target preliminary studies confirmed the hypothesis that this new class of compounds inhibits Trp biosynthesis. Taken together, these findings indicate that fluorophenylbenzohydrazides represent good candidates to be assessed for drug discovery.
Collapse
Affiliation(s)
- Sara Consalvi
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Rome, Italy
| | - Giulia Venditti
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Rome, Italy
| | - Junhao Zhu
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Helena I Boshoff
- National Institute of Allergy and Infectious Diseases, Laboratory of Clinical Immunology and Microbiology, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Kriti Arora
- National Institute of Allergy and Infectious Diseases, Laboratory of Clinical Immunology and Microbiology, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Alessandro De Logu
- Department of Life and Environmental Sciences, University of Cagliari, via Ospedale 72, 09124, Cagliari, Italy
| | - Thomas R Ioerger
- Department of Computer Science, Texas A&M University, 3112 TAMU, College Station, TX, 77843, USA
| | - Eric J Rubin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Mariangela Biava
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Rome, Italy.
| | - Giovanna Poce
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Rome, Italy.
| |
Collapse
|
42
|
Jiang J, Li Z, Chen C, Jiang W, Xu B, Zhao Q. Metabolomics Strategy Assisted by Transcriptomics Analysis to Identify Potential Biomarkers Associated with Tuberculosis. Infect Drug Resist 2021; 14:4795-4807. [PMID: 34815677 PMCID: PMC8604652 DOI: 10.2147/idr.s330493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/28/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose To investigate the dysregulated pathways and identify reliable diagnostic biomarkers for tuberculosis using integrated analysis of metabolomics and transcriptomics. Methods Three groups of samples, untargeted metabolomics analysis of healthy controls (HC), latent tuberculosis infection patients (LTBI), and active tuberculosis patients (TB), were analyzed using gas chromatography time-of-flight mass spectrometry (GC-TOF MS) and ultra-high performance liquid chromatography-quantitative mass spectrometry (UHPLC-QE-MS). Both univariate and multivariate and statistical analyses were used to select differential metabolites (DMs) among group comparison, and LASSO regression analysis was employed to discover potential diagnostic biomarkers. Metabolite set enrichment analysis was performed to identify the altered metabolic pathways specifically in patients with TB. Meanwhile, a transcriptomic dataset GSEG4992 was downloaded from the GEO database to explore the differentially expressed genes (DEGs) between TB and HC identified in significantly enriched pathways. Finally, an integrative analysis of DMs and DEGs was performed to investigate the possible molecular mechanisms of TB. Results Thirty-three specific metabolites were significantly different between TB and HC, of which 7 (5-hydroxyindoleacetic acid, isoleucyl-isoleucine, heptadecanoic acid, indole acetaldehyde, 5-ethyl-2,4-dimethyloxazole, and 2-hydroxycaproic acid, unknown 71) were chosen as combinational potential biomarkers for TB. The area under the curve (AUC) value of these biomarkers was 0.97 (95% CI: 0.92–1.00). Metabolites set enrichment analysis (MSEA) displayed that there were 3 significantly enriched pathways among all. The genes in 3 significantly enriched pathways were further analyzed, of which 9(ALDH3B1, BCAT1, BCAT2, GLYAT, GOT1, IL4I1, MIF, SDS, SDSL) were expressed differentially. The area under the curve (AUC) values of these DEGs enriched in pathways mostly were greater than 0.8. As a result, a connected network of metabolites and genes in the pathways were established, which provides insights into the credibility of selected metabolites. Conclusion The newly identified metabolic biomarkers display a high potential to be developed into a promising tool for TB screening, diagnosis, and therapeutic effect monitoring.
Collapse
Affiliation(s)
- Jiayan Jiang
- School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, People's Republic of China
| | - Zhipeng Li
- School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, People's Republic of China
| | - Cheng Chen
- Department of Chronic Communicable Disease, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Weili Jiang
- School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, People's Republic of China
| | - Biao Xu
- School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, People's Republic of China
| | - Qi Zhao
- School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, People's Republic of China.,NHC Key Laboratory of Health Technology Assessment,Fudan University, Shanghai, People's Republic of China.,Shanghai Clinical Research Center for infectious disease (Tuberculosis), Shanghai, People's Republic of China
| |
Collapse
|
43
|
Borah K, Xu Y, McFadden J. Dissecting Host-Pathogen Interactions in TB Using Systems-Based Omic Approaches. Front Immunol 2021; 12:762315. [PMID: 34795672 PMCID: PMC8593131 DOI: 10.3389/fimmu.2021.762315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/18/2021] [Indexed: 01/10/2023] Open
Abstract
Tuberculosis (TB) is a devastating infectious disease that kills over a million people every year. There is an increasing burden of multi drug resistance (MDR) and extensively drug resistance (XDR) TB. New and improved therapies are urgently needed to overcome the limitations of current treatment. The causative agent, Mycobacterium tuberculosis (Mtb) is one of the most successful pathogens that can manipulate host cell environment for adaptation, evading immune defences, virulence, and pathogenesis of TB infection. Host-pathogen interaction is important to establish infection and it involves a complex set of processes. Metabolic cross talk between the host and pathogen is a facet of TB infection and has been an important topic of research where there is growing interest in developing therapies and drugs that target these interactions and metabolism of the pathogen in the host. Mtb scavenges multiple nutrient sources from the host and has adapted its metabolism to survive in the intracellular niche. Advancements in systems-based omic technologies have been successful to unravel host-pathogen interactions in TB. In this review we discuss the application and usefulness of omics in TB research that provides promising interventions for developing anti-TB therapies.
Collapse
Affiliation(s)
- Khushboo Borah
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | | | - Johnjoe McFadden
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
44
|
Liebenberg C, Luies L, Williams AA. Metabolomics as a Tool to Investigate HIV/TB Co-Infection. Front Mol Biosci 2021; 8:692823. [PMID: 34746228 PMCID: PMC8565463 DOI: 10.3389/fmolb.2021.692823] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 10/04/2021] [Indexed: 12/28/2022] Open
Abstract
The HIV/AIDS (human immunodeficiency virus/acquired immunodeficiency syndrome) and tuberculosis (TB) pandemics are perpetuated by a significant global burden of HIV/TB co-infection. The synergy between HIV and Mycobacterium tuberculosis (Mtb) during co-infection of a host is well established. While this synergy is known to be driven by immunological deterioration, the metabolic mechanisms thereof remain poorly understood. Metabolomics has been applied to study various aspects of HIV and Mtb infection separately, yielding insights into infection- and treatment-induced metabolic adaptations experienced by the host. Despite the contributions that metabolomics has made to the field, this approach has not yet been systematically applied to characterize the HIV/TB co-infected state. Considering that limited HIV/TB co-infection metabolomics studies have been published to date, this review briefly summarizes what is known regarding the HIV/TB co-infection synergism from a conventional and metabolomics perspective. It then explores metabolomics as a tool for the improved characterization of HIV/TB co-infection in the context of previously published human-related HIV infection and TB investigations, respectively as well as for addressing the gaps in existing knowledge based on the similarities and deviating trends reported in these HIV infection and TB studies.
Collapse
|
45
|
TCA cycle remodeling drives proinflammatory signaling in humans with pulmonary tuberculosis. PLoS Pathog 2021; 17:e1009941. [PMID: 34559866 PMCID: PMC8494353 DOI: 10.1371/journal.ppat.1009941] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/06/2021] [Accepted: 09/03/2021] [Indexed: 11/19/2022] Open
Abstract
The metabolic signaling pathways that drive pathologic tissue inflammation and damage in humans with pulmonary tuberculosis (TB) are not well understood. Using combined methods in plasma high-resolution metabolomics, lipidomics and cytokine profiling from a multicohort study of humans with pulmonary TB disease, we discovered that IL-1β-mediated inflammatory signaling was closely associated with TCA cycle remodeling, characterized by accumulation of the proinflammatory metabolite succinate and decreased concentrations of the anti-inflammatory metabolite itaconate. This inflammatory metabolic response was particularly active in persons with multidrug-resistant (MDR)-TB that received at least 2 months of ineffective treatment and was only reversed after 1 year of appropriate anti-TB chemotherapy. Both succinate and IL-1β were significantly associated with proinflammatory lipid signaling, including increases in the products of phospholipase A2, increased arachidonic acid formation, and metabolism of arachidonic acid to proinflammatory eicosanoids. Together, these results indicate that decreased itaconate and accumulation of succinate and other TCA cycle intermediates is associated with IL-1β-mediated proinflammatory eicosanoid signaling in pulmonary TB disease. These findings support host metabolic remodeling as a key driver of pathologic inflammation in human TB disease. Pulmonary tuberculosis (TB) often results in pathologic lung inflammation that causes tissue damage and does not control bacterial replication. This impairs the host response to antibiotic treatment and can result in long term deficits in lung function. Currently, the role of host metabolism in regulating the inflammatory response in TB disease is not well understood. Here, we use detailed immunometabolic phenotyping to show that metabolic remodeling of the tricarboxylic acid (TCA) cycle is closely associated with pathologic inflammatory signaling in humans with TB disease. Accumulation of TCA cycle intermediates in plasma, including the proinflammatory metabolite succinate, as well as decreased concentrations of the anti-inflammatory metabolite itaconate, were associated with increases in IL-1β and upregulation of proinflammatory lipid signaling cascades. This inflammatory network was upregulated following delays in appropriate anti-TB treatment and was associated with prolonged time to sputum culture clearance of Mycobacterium tuberculosis. Our study provides new insights into the metabolic reprograming that leads to pathologic inflammation in humans with pulmonary TB.
Collapse
|
46
|
Marszalek-Grabska M, Walczak K, Gawel K, Wicha-Komsta K, Wnorowska S, Wnorowski A, Turski WA. Kynurenine emerges from the shadows – Current knowledge on its fate and function. Pharmacol Ther 2021; 225:107845. [DOI: 10.1016/j.pharmthera.2021.107845] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022]
|
47
|
An antitubercular prodrug leaves Mycobacterium tuberculosis facing a difficult choice, poisoning or starvation? Cell Chem Biol 2021; 28:1101-1103. [PMID: 34416139 DOI: 10.1016/j.chembiol.2021.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this issue of Cell Chemical Biology, Libardo et al. (2021) identify prodrugs that kill Mtb through poisoning of its L-tryptophan biosynthetic pathway. Determination of the mechanisms of resistance evolved by the bacterium highlights the importance of metabolic flux modulation in TB drug resistance.
Collapse
|
48
|
Wang X, Mehra S, Kaushal D, Veazey RS, Xu H. Abnormal Tryptophan Metabolism in HIV and Mycobacterium tuberculosis Infection. Front Microbiol 2021; 12:666227. [PMID: 34262540 PMCID: PMC8273495 DOI: 10.3389/fmicb.2021.666227] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Host metabolism has recently gained more attention for its roles in physiological functions and pathologic conditions. Of these, metabolic tryptophan disorders generate a pattern of abnormal metabolites that are implicated in various diseases. Here, we briefly highlight the recent advances regarding abnormal tryptophan metabolism in HIV and Mycobacterium tuberculosis infection and discuss its potential impact on immune regulation, disease progression, and neurological disorders. Finally, we also discuss the potential for metabolic tryptophan interventions toward these infectious diseases.
Collapse
Affiliation(s)
- Xiaolei Wang
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, United States
| | - Smriti Mehra
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, United States
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Ronald S. Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, United States
| | - Huanbin Xu
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, United States
| |
Collapse
|
49
|
Schnittman SR, Deitchman AN, Beck-Engeser G, Ahn H, York VA, Hartig H, Hecht FM, Martin JN, Deeks SG, Aweeka FT, Hunt PW. Abnormal Levels of Some Biomarkers of Immune Activation Despite Very Early Treatment of Human Immunodeficiency Virus. J Infect Dis 2021; 223:1621-1630. [PMID: 32915986 PMCID: PMC8136975 DOI: 10.1093/infdis/jiaa580] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Despite early antiretroviral therapy (ART), ART-suppressed people with human immunodeficiency virus (HIV) (PWH) remain at higher risk for infections and infection-related cancers than the general population. The immunologic pathways that remain abnormal in this setting, potentially contributing to these complications, are unclear. METHODS ART-suppressed PWH and HIV-negative controls, all cytomegalovirus seropositive and enriched for HIV risk factors, were sampled from an influenza vaccine responsiveness study. PWH were stratified by timing of ART initiation (within 6 months of infection [early ART] vs later) and nadir CD4+ T-cell count among later initiators. Between-group differences in kynurenine-tryptophan (KT) ratio, interferon-inducible protein 10, soluble CD14 and CD163, soluble tumor necrosis factor receptor 2, interleukin 6, and soluble urokinase plasminogen activator receptor were assessed after confounder adjustment. RESULTS Most participants (92%) were male, reflecting the demographics of early-ART initiators in San Francisco. Most biomarkers were higher among later-ART initiators. Participants in the early-ART group achieved near-normal soluble tumor necrosis factor receptor 2, interleukin 6, and soluble urokinase plasminogen activator receptor levels, but substantially higher KT ratio than those without HIV after confounder adjustment (P = .008). Soluble CD14, soluble CD163, and interferon-inducible protein 10 trended similarly. CONCLUSIONS While early-ART initiators restore near-normal levels of many inflammatory markers, the kynurenine pathway of tryptophan catabolism remains abnormally high. Because this pathway confers adaptive immune defects and predicts tuberculosis and cancer progression, this it may contribute to persistent risks of these complications in this setting.
Collapse
Affiliation(s)
- Samuel R Schnittman
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Amelia N Deitchman
- Department of Clinical Pharmacy, School of Pharmacy, University of California, San Francisco, California, USA
| | - Gabriele Beck-Engeser
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - HaeLee Ahn
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Vanessa A York
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Heather Hartig
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Frederick M Hecht
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey N Martin
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, California, USA
| | - Steven G Deeks
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Francesca T Aweeka
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Department of Clinical Pharmacy, School of Pharmacy, University of California, San Francisco, California, USA
| | - Peter W Hunt
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
50
|
Schnittman SR, Byakwaga H, Boum Y, Kabakyenga J, Matthews LT, Burdo TH, Huang Y, Tracy RP, Haberer JE, Kembabazi A, Kaida A, Moisi D, Lederman MM, Bangsberg DR, Martin JN, Hunt PW. Changes in Immune Activation During Pregnancy and the Postpartum Period in Treated HIV Infection. Open Forum Infect Dis 2021; 8:ofab245. [PMID: 34159218 PMCID: PMC8214017 DOI: 10.1093/ofid/ofab245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/12/2021] [Indexed: 12/30/2022] Open
Abstract
Background Pregnant women with HIV (PWWH) have high postpartum morbidity and mortality from infections like tuberculosis. Immunologic changes during pregnancy and postpartum periods may contribute to these risks, particularly the immunoregulatory kynurenine pathway of tryptophan catabolism, which contributes to both HIV and tuberculosis pathogenesis and increases in the early postpartum period. Methods Women with HIV initiating antiretroviral therapy (ART) in the Uganda AIDS Rural Treatment Outcomes (UARTO) cohort who were pregnant at enrollment or became pregnant during observation were studied (n = 54). Plasma kynurenine/tryptophan (KT) ratio, soluble CD14 (sCD14), sCD163, sCD27, interferon-inducible protein 10 (IP-10), D-dimer, interleukin-6, and intestinal fatty-acid binding protein levels were assessed through the first year of ART and at 3-month intervals throughout pregnancy and 1 year postpartum. Biomarker changes were assessed with linear mixed models adjusted for ART duration. Hemoglobin concentration changes were used to estimate pregnancy-related changes in plasma volume. Results The median pre-ART CD4 count was 134. D-dimer increased through the third trimester before returning to baseline postpartum, while most other biomarkers declined significantly during pregnancy, beyond what would be expected from pregnancy-associated plasma volume expansion. IP-10 and sCD14 remained suppressed for at least 12 months postpartum. KT ratio was the only biomarker that increased above prepregnancy baseline postpartum (mean + 30%; P < .001) and remained higher than baseline for ≥9 months (P ≤ .045 for all time points). Conclusions Several immune activation markers decline during pregnancy and remain suppressed postpartum, but the kynurenine pathway of tryptophan catabolism increases above baseline for ≥9 months postpartum. The mechanisms underlying postpartum kynurenine pathway activity are incompletely understood but may contribute to increased tuberculosis risk in this setting.
Collapse
Affiliation(s)
| | - Helen Byakwaga
- Mbarara University of Science and Technology, Mbarara, Uganda
| | - Yap Boum
- Mbarara University of Science and Technology, Mbarara, Uganda
| | | | - Lynn T Matthews
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Tricia H Burdo
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Yong Huang
- University of California San Francisco, San Francisco, California, USA
| | | | - Jessica E Haberer
- Massachusetts General Hospital and Harvard University, Boston, Massachusetts, USA
| | - Annet Kembabazi
- Mbarara University of Science and Technology, Mbarara, Uganda
| | - Angela Kaida
- Simon Fraser University, Vancouver, British Columbia, Canada
| | - Daniela Moisi
- Case Western Reserve University, Cleveland, Ohio, USA
| | | | - David R Bangsberg
- Mbarara University of Science and Technology, Mbarara, Uganda.,Oregon Health and Sciences University and Portland State University School of Public Health, Portland, Oregon, USA
| | - Jeffrey N Martin
- University of California San Francisco, San Francisco, California, USA
| | - Peter W Hunt
- University of California San Francisco, San Francisco, California, USA
| |
Collapse
|