1
|
Malik AA, Shariq M, Sheikh JA, Fayaz H, Srivastava G, Thakuri D, Ahuja Y, Ali S, Alam A, Ehtesham NZ, Hasnain SE. Regulation of Type I Interferon and Autophagy in Immunity against Mycobacterium Tuberculosis: Role of CGAS and STING1. Adv Biol (Weinh) 2024; 8:e2400174. [PMID: 38977406 DOI: 10.1002/adbi.202400174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/22/2024] [Indexed: 07/10/2024]
Abstract
Mycobacterium tuberculosis (M. tb) is a significant intracellular pathogen responsible for numerous infectious disease-related deaths worldwide. It uses ESX-1 T7SS to damage phagosomes and to enter the cytosol of host cells after phagocytosis. During infection, M. tb and host mitochondria release dsDNA, which activates the CGAS-STING1 pathway. This pathway leads to the production of type I interferons and proinflammatory cytokines and activates autophagy, which targets and degrades bacteria within autophagosomes. However, the role of type I IFNs in immunity against M. tb is controversial. While previous research has suggested a protective role, recent findings from cgas-sting1 knockout mouse studies have contradicted this. Additionally, a study using knockout mice and non-human primate models uncovered a new mechanism by which neutrophils recruited to lung infections form neutrophil extracellular traps. Activating plasmacytoid dendritic cells causes them to produce type I IFNs, which interfere with the function of interstitial macrophages and increase the likelihood of tuberculosis. Notably, M. tb uses its virulence proteins to disrupt the CGAS-STING1 signaling pathway leading to enhanced pathogenesis. Investigating the CGAS-STING1 pathway can help develop new ways to fight tuberculosis.
Collapse
Affiliation(s)
- Asrar Ahmad Malik
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Mohd Shariq
- ICMR-National Institute of Pathology, Ansari Nagar West, New Delhi, 110029, India
| | - Javaid Ahmad Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Haleema Fayaz
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Gauri Srivastava
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Deeksha Thakuri
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Yashika Ahuja
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Saquib Ali
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Anwar Alam
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Nasreen Z Ehtesham
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Seyed E Hasnain
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), Hauz Khas, New Delhi, 110 016, India
| |
Collapse
|
2
|
Perez RL, Chase J, Tanner R. Shared challenges to the control of complex intracellular neglected pathogens. Front Public Health 2024; 12:1423420. [PMID: 39324165 PMCID: PMC11422159 DOI: 10.3389/fpubh.2024.1423420] [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: 04/25/2024] [Accepted: 08/13/2024] [Indexed: 09/27/2024] Open
Abstract
The complex intracellular pathogens Mycobacterium tuberculosis, Mycobacterium leprae, Leishmania spp., and Burkholderia pseudomallei, which cause tuberculosis, leprosy, leishmaniasis, and melioidosis respectively, represent major health threats with a significant global burden concentrated in low- and middle-income countries. While these diseases vary in their aetiology, pathology and epidemiology, they share key similarities in the biological and sociodemographic factors influencing their incidence and impact worldwide. In particular, their occurrence in resource-limited settings has important implications for research and development, disease prevalence and associated risk factors, as well as access to diagnostics and therapeutics. In accordance with the vision of the VALIDATE (VAccine deveLopment for complex Intracellular neglecteD pAThogeEns) Network, we consider shared challenges to the effective prevention, diagnosis and treatment of these diseases as shaped by both biological and social factors, illustrating the importance of taking an interdisciplinary approach. We further highlight how a cross-pathogen perspective may provide valuable insights for understanding and addressing challenges to the control of all four pathogens.
Collapse
Affiliation(s)
- Rebecca Lynn Perez
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Wadham College, University of Oxford, Oxford, United Kingdom
| | - Jemima Chase
- Wadham College, University of Oxford, Oxford, United Kingdom
| | - Rachel Tanner
- Wadham College, University of Oxford, Oxford, United Kingdom
- Department of Biology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
3
|
Randall EM, Sopp P, Raper A, Dry I, Burdon T, Hope JC, Waddell LA. Phenotypic characterisation of bovine alveolar macrophages reveals two major subsets with differential expression of CD163. Sci Rep 2024; 14:14974. [PMID: 38951667 PMCID: PMC11217380 DOI: 10.1038/s41598-024-65868-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Bovine alveolar macrophages (AMs) defend the lungs against pathogens such as Mycobacterium bovis (M. bovis), the causative agent of bovine tuberculosis. However, little is known about the surface molecules expressed by bovine AMs and whether there is heterogeneity within the population. The purpose of this study was to characterise the bovine AM cell surface phenotype using flow cytometry. Bronchoalveolar lavage samples from four different calves were stained with a combination of antibodies against immune cell molecules prior to flow cytometric analysis. To assess the degree of expression, we considered the distribution and relative intensities of stained and unstained cells. We demonstrated that bovine AMs have high expression of CD172a, ADGRE1, CD206, and CD14, moderate expression of CD80, MHC II, CD1b, and CD40, low expression of CX3CR1 and CD86, and little or no expression of CD16 and CD26. Two distinct subsets of bovine AMs were identified based on CD163 expression. Subsequent analysis showed that the CD163+ subset had greater expression of other typical macrophage molecules compared to the CD163- subset, suggesting that these cells may perform different roles during infection. The characterisation of the uninfected bovine AM phenotype will provide a foundation for the examination of M. bovis-infected AMs.
Collapse
Affiliation(s)
| | - Paul Sopp
- MRC Weatherall Institute for Molecular Medicine, John Radcliffe Hospital, Oxford, OX39DS, UK
| | - Anna Raper
- The Roslin Institute, Easter Bush, Edinburgh, EH25 9RG, UK
| | - Inga Dry
- The Roslin Institute, Easter Bush, Edinburgh, EH25 9RG, UK
| | - Tom Burdon
- The Roslin Institute, Easter Bush, Edinburgh, EH25 9RG, UK
| | - Jayne C Hope
- The Roslin Institute, Easter Bush, Edinburgh, EH25 9RG, UK.
| | | |
Collapse
|
4
|
Breen P, Zimbric M, Caverly LJ. Itaconic acid inhibits nontuberculous mycobacterial growth in pH dependent manner while 4-octyl-itaconic acid enhances THP-1 clearance of nontuberculous mycobacteria in vitro. PLoS One 2024; 19:e0303516. [PMID: 38728330 PMCID: PMC11086914 DOI: 10.1371/journal.pone.0303516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
Increasingly prevalent, nontuberculous mycobacteria (NTM) infections affect approximately 20% of people with cystic fibrosis (CF). Previous studies of CF sputum identified lower levels of the host metabolite itaconate in those infected with NTM. Itaconate can inhibit the growth of M. tuberculosis (MTB) in vitro via the inhibition of the glyoxylate cycle enzyme (ICL), but its impact on NTM is unclear. To test itaconic acid's (IA) effect on NTM growth, laboratory and CF clinical strains of Mycobacterium abscessus and Mycobacterium avium were cultured in 7H9 minimal media supplemented with 1-10 mM of IA and short-chain fatty acids (SCFA). M. avium and M. abscessus grew when supplemented with SCFAs, whereas the addition of IA (≥ 10 mM) completely inhibited NTM growth. NTM supplemented with acetate or propionate and 5 mM IA displayed slower growth than NTM cultured with SCFA and ≤ 1 mM of IA. However, IA's inhibition of NTM was pH dependent; as similar and higher quantities (100 mM) of pH adjusted IA (pH 7) did not inhibit growth in vitro, while in an acidic minimal media (pH 6.1), 1 to 5 mM of non-pH adjusted IA inhibited growth. None of the examined isolates displayed the ability to utilize IA as a carbon source, and IA added to M. abscessus isocitrate lyase (ICL) decreased enzymatic activity. Lastly, the addition of cell-permeable 4-octyl itaconate (4-OI) to THP-1 cells enhanced NTM clearance, demonstrating a potential role for IA/itaconate in host defense against NTM infections.
Collapse
Affiliation(s)
- Paul Breen
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Madsen Zimbric
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Lindsay J. Caverly
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States of America
| |
Collapse
|
5
|
Malik AA, Shariq M, Sheikh JA, Zarin S, Ahuja Y, Fayaz H, Alam A, Ehtesham NZ, Hasnain SE. Activation of the lysosomal damage response and selective autophagy: the coordinated actions of galectins, TRIM proteins, and CGAS-STING1 in providing immunity against Mycobacterium tuberculosis. Crit Rev Microbiol 2024:1-20. [PMID: 38470107 DOI: 10.1080/1040841x.2024.2321494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 02/14/2024] [Indexed: 03/13/2024]
Abstract
Autophagy is a crucial immune defense mechanism that controls the survival and pathogenesis of M. tb by maintaining cell physiology during stress and pathogen attack. The E3-Ub ligases (PRKN, SMURF1, and NEDD4) and autophagy receptors (SQSTM1, TAX1BP1, CALCOCO2, OPTN, and NBR1) play key roles in this process. Galectins (LGALSs), which bind to sugars and are involved in identifying damaged cell membranes caused by intracellular pathogens such as M. tb, are essential. These include LGALS3, LGALS8, and LGALS9, which respond to endomembrane damage and regulate endomembrane damage caused by toxic chemicals, protein aggregates, and intracellular pathogens, including M. tb. They also activate selective autophagy and de novo endolysosome biogenesis. LGALS3, LGALS9, and LGALS8 interact with various components to activate autophagy and repair damage, while CGAS-STING1 plays a critical role in providing immunity against M. tb by activating selective autophagy and producing type I IFNs with antimycobacterial functions. STING1 activates cGAMP-dependent autophagy which provides immunity against various pathogens. Additionally, cytoplasmic surveillance pathways activated by ds-DNA, such as inflammasomes mediated by NLRP3 and AIM2 complexes, control M. tb. Modulation of E3-Ub ligases with small regulatory molecules of LGALSs and TRIM proteins could be a novel host-based therapeutic approach for controlling TB.
Collapse
Affiliation(s)
- Asrar Ahmad Malik
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Mohd Shariq
- ICMR-National Institute of Pathology, New Delhi, India
| | - Javaid Ahmad Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, New Delhi, India
| | - Sheeba Zarin
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, New Delhi, India
| | - Yashika Ahuja
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Haleema Fayaz
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Anwar Alam
- Department of Biotechnology, School of Science and Engineering Technology, Sharda University, Greater Noida, India
| | - Nasreen Z Ehtesham
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Seyed E Hasnain
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| |
Collapse
|
6
|
Samuels V, Mulelu AE, Ndlovu H, Marakalala MJ. Mycobacterial FtsEX-RipC interaction is required for normal growth and cell morphology in rifampicin and low ionic strength conditions. Microbiol Spectr 2024; 12:e0251523. [PMID: 38289931 PMCID: PMC10913748 DOI: 10.1128/spectrum.02515-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
Tuberculosis, a lung disease caused by Mycobacterium tuberculosis (Mtb), remains a major global health problem ranking as the second leading cause of death from a single infectious agent. One of the major factors contributing toward Mtb's success as a pathogen is its unique cell wall and its ability to counteract various arms of the host's immune response. A recent genome-scale study profiled a list of candidate genes that are predicted to be essential for Mtb survival of host-mediated responses. One candidate was FtsEX, a protein complex composed of an ATP-binding domain, FtsE, and a transmembrane domain, FtsX. FtsEX functions through interaction with a periplasmic hydrolase, RipC. Homologs of FtsEX exist in other bacteria and have been linked with playing a key role in regulating peptidoglycan hydrolysis during cell elongation and division. Here, we report on Mycobacterium smegmatis, FtsE, FtsX, and RipC and their protective roles in stressful conditions. We demonstrate that the individual genes of FtsEX complex and RipC are not essential for survival in normal growth conditions but conditionally essential in low-salt media and antibiotic-treated media. Growth defects in these conditions were characterized by short and bulgy cells as well as elongated filamentous cells. Our results suggest that FtsE, FtsX, and RipC are required for both normal cell elongation and division and ultimately for survival in stressful conditions. IMPORTANCE Mycobacterial cell growth and division are coordinated with regulated peptidoglycan hydrolysis. Understanding cell wall gene complexes that govern normal cell division and elongation will aid in the development of tools to disarm the ability of mycobacteria to survive immune-like and antibiotic stresses. We combined genetic analyses and scanning electron microscopy to analyze morphological changes of mycobacterial FtsEX and RipC mutants in stressful conditions. We demonstrate that FtsE, FtsX, FtsEX, and RipC are conditionally required for the survival of Mycobacterium smegmatis during rifampicin treatment and in low-salt conditions. Growth defects in these conditions were characterized by short and bulgy cells as well as elongated filamentous cells. We also show that the FtsEX-RipC interaction is essential for the survival of M. smegmatis in rifampicin. Our results suggest that FtsE, FtsX, and RipC are required for normal cell wall regulation and ultimately for survival in stressful conditions.
Collapse
Affiliation(s)
- Veneshley Samuels
- Division of Medical Microbiology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Andani E. Mulelu
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Hlumani Ndlovu
- Division of Chemical Systems Biology, Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mohlopheni J. Marakalala
- Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Africa Health Research Institute, Durban, KwaZulu-Natal, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
7
|
Manabile MA, Maguga-Phasha TC, Makgatho ME. Effects of SNPs on TNF-α and IL-10 cytokine expression in TB and HIV patients in the Capricorn district, Limpopo Province, South Africa. Afr Health Sci 2024; 24:1-9. [PMID: 38962334 PMCID: PMC11217856 DOI: 10.4314/ahs.v24i1.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024] Open
Abstract
Background The impact of Tuberculosis (TB) places an immense burden on the health care system. Infection with Human Immunodeficiency Virus (HIV) is a significant risk factor in the development and progression of TB disease. Single Nucleotide Polymorphisms (SNPs) in the promoter region of Interleukin-10 (IL-10) and Tumour Necrotic Factor-Alpha (TNF-α) may play a major role in the disease mechanism and understanding these mechanisms might prove to be a useful diagnostic tool in evaluating the immune regulation and progression of the disease. Objective This study aimed to determine the relationship between cytokine levels and gene variants of Interleukin-10 and Tumour Necrotic Factor Alpha in TB and HIV-infected participants. Methods Cytokine levels were determined by ELISA, and SNPs were determined by MassArray®. Results The levels of TNF-α were higher in the TB group than the HIV (p < 0.001) and TB-HIV (p = 0.011) groups, but similar to the TNF-α levels in the control group. In the HIV group, IL-10 levels were higher than those of the TB (p < 0.001) and control groups (p = 0.039), whereas there was no difference between the IL-10 levels in the HIV and the TB-HIV infection groups. The ratio was determined and there were no differences between the four infection groups. In this study, no associations were detected between the circulating plasma levels of TNF-α and IL-10 and their genotypes. Conclusion Our data showed that the gene variants were not associated with circulating plasma levels of TNF-α and IL-10 in our study population. A pro-inflammatory environment was found in the TB and TB-HIV groups, which is suggesting of bacterial clearance, while an anti-inflammatory environment was found in the HIV group, which suggests the suppression of viral replication.
Collapse
Affiliation(s)
- Mosebo A Manabile
- Pathology and Medical Sciences, University of Limpopo, Limpopo Province, South Africa
| | | | - Marema E Makgatho
- Pathology and Medical Sciences, University of Limpopo, Limpopo Province, South Africa
| |
Collapse
|
8
|
Cupido G, Günther G. Post tuberculosis lung disease and tuberculosis sequelae: A narrative review. Indian J Tuberc 2024; 71:64-72. [PMID: 38296392 DOI: 10.1016/j.ijtb.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 02/08/2024]
Abstract
Post Tuberculosis lung disease (PTLD) and post tuberculosis sequelae is a global and poorly recognized problem, amplified by social factors and immunocompromising conditions, inadequate treatment, lack of effective prevention of tuberculosis (TB) infection and disease. As a disease, it remained until recently poorly defined, with studies heterogenous with regards to regions, population demographics, risk factors, cohort sizes, and methods. Pathophysiologically, even successfully treated pulmonary TB disease has sequelae i.e. involving central and peripheral airways, lung parenchyma and pleura, resulting in airway narrowing and dilatation, fibrocavitation and emphysema, pulmonary vascular changes as well as pleural fibrosis. Functionally patients have airflow limitation, restrictive disease or a mixture of both not rarely associated with respiratory, or even ventilatory failure. Quality of life is often impaired through disability, TB relapse, superinfections and through increased susceptibility to reinfection and persistent inflammation, leading to progressive lung function decline and an increased risk of cardiovascular disease and cancer. Premature mortality due to PTLD is very likely, but poorly described.
Collapse
Affiliation(s)
- Gordon Cupido
- Department of Internal Medicine, Katutura State Hospital, Windhoek, Namibia.
| | - Gunar Günther
- Department of Pulmonology and Allergology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Medical Sciences, University of Namibia, School of Medicine, Windhoek, Namibia
| |
Collapse
|
9
|
Steele MI, Peiser JM, Shreenidhi PM, Strassmann JE, Queller DC. Predation-resistant Pseudomonas bacteria engage in symbiont-like behavior with the social amoeba Dictyostelium discoideum. THE ISME JOURNAL 2023; 17:2352-2361. [PMID: 37884792 PMCID: PMC10689837 DOI: 10.1038/s41396-023-01535-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023]
Abstract
The soil amoeba Dictyostelium discoideum acts as both a predator and potential host for diverse bacteria. We tested fifteen Pseudomonas strains that were isolated from transiently infected wild D. discoideum for ability to escape predation and infect D. discoideum fruiting bodies. Three predation-resistant strains frequently caused extracellular infections of fruiting bodies but were not found within spores. Furthermore, infection by one of these species induces secondary infections and suppresses predation of otherwise edible bacteria. Another strain can persist inside of amoebae after being phagocytosed but is rarely taken up. We sequenced isolate genomes and discovered that predation-resistant isolates are not monophyletic. Many Pseudomonas isolates encode secretion systems and toxins known to improve resistance to phagocytosis in other species, as well as diverse secondary metabolite biosynthetic gene clusters that may contribute to predation resistance. However, the distribution of these genes alone cannot explain why some strains are edible and others are not. Each lineage may employ a unique mechanism for resistance.
Collapse
Affiliation(s)
- Margaret I Steele
- Biology Department, Washington University in St. Louis, St. Louis, MO, USA.
| | - Jessica M Peiser
- Biology Department, Washington University in St. Louis, St. Louis, MO, USA
| | - P M Shreenidhi
- Biology Department, Washington University in St. Louis, St. Louis, MO, USA
| | - Joan E Strassmann
- Biology Department, Washington University in St. Louis, St. Louis, MO, USA
| | - David C Queller
- Biology Department, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
10
|
Kayongo A, Nyiro B, Siddharthan T, Kirenga B, Checkley W, Lutaakome Joloba M, Ellner J, Salgame P. Mechanisms of lung damage in tuberculosis: implications for chronic obstructive pulmonary disease. Front Cell Infect Microbiol 2023; 13:1146571. [PMID: 37415827 PMCID: PMC10320222 DOI: 10.3389/fcimb.2023.1146571] [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: 01/17/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
Pulmonary tuberculosis is increasingly recognized as a risk factor for COPD. Severe lung function impairment has been reported in post-TB patients. Despite increasing evidence to support the association between TB and COPD, only a few studies describe the immunological basis of COPD among TB patients following successful treatment completion. In this review, we draw on well-elaborated Mycobacterium tuberculosis-induced immune mechanisms in the lungs to highlight shared mechanisms for COPD pathogenesis in the setting of tuberculosis disease. We further examine how such mechanisms could be exploited to guide COPD therapeutics.
Collapse
Affiliation(s)
- Alex Kayongo
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Brian Nyiro
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Trishul Siddharthan
- Division of Pulmonary and Critical Care Medicine, University of Miami, Miami, FL, United States
| | - Bruce Kirenga
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - William Checkley
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
- Center for Global Non-Communicable Disease Research and Training, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Moses Lutaakome Joloba
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Jerrold Ellner
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Padmini Salgame
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
11
|
Fernandes GFS, Manieri KF, Bonjorno AF, Campos DL, Ribeiro CM, Demarqui FM, Ruiz DAG, Nascimento-Junior NM, Denny WA, Thompson AM, Pavan FR, Dos Santos JL. Synthesis and Anti-Mycobacterium tuberculosis Activity of Imidazo[2,1-b][1,3]oxazine Derivatives against Multidrug-Resistant Strains. ChemMedChem 2023; 18:e202300015. [PMID: 37002895 DOI: 10.1002/cmdc.202300015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/09/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023]
Abstract
The emergence of multidrug-resistant strains of M. tuberculosis has raised concerns due to the greater difficulties in patient treatment and higher mortality rates. Herein, we revisited the 2-nitro-6,7-dihydro-5H-imidazo[2,1-b][1,3]oxazine scaffold and identified potent new carbamate derivatives having MIC90 values of 0.18-1.63 μM against Mtb H37Rv. Compounds 47-49, 51-53, and 55 exhibited remarkable activity against a panel of clinical isolates, displaying MIC90 values below 0.5 μM. In Mtb-infected macrophages, several compounds demonstrated a 1-log greater reduction in mycobacterial burden than rifampicin and pretomanid. The compounds tested did not exhibit significant cytotoxicity against three cell lines or any toxicity to Galleria mellonella. Furthermore, the imidazo[2,1-b][1,3]oxazine derivatives did not show substantial activity against other bacteria or fungi. Finally, molecular docking studies revealed that the new compounds could interact with the deazaflavin-dependent nitroreductase (Ddn) in a similar manner to pretomanid. Collectively, our findings highlight the chemical universe of imidazo[2,1-b][1,3]oxazines and their promising potential against MDR-TB.
Collapse
Affiliation(s)
- Guilherme F S Fernandes
- School of Pharmaceutical Sciences, São Paulo State University, Rod. Araraquara-Jaú, Araraquara, 14800903, Brazil
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Present address: Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, UK
| | - Karyn F Manieri
- School of Pharmaceutical Sciences, São Paulo State University, Rod. Araraquara-Jaú, Araraquara, 14800903, Brazil
| | - Andressa F Bonjorno
- School of Pharmaceutical Sciences, São Paulo State University, Rod. Araraquara-Jaú, Araraquara, 14800903, Brazil
| | - Debora L Campos
- School of Pharmaceutical Sciences, São Paulo State University, Rod. Araraquara-Jaú, Araraquara, 14800903, Brazil
| | - Camila M Ribeiro
- School of Pharmaceutical Sciences, São Paulo State University, Rod. Araraquara-Jaú, Araraquara, 14800903, Brazil
| | - Fernanda M Demarqui
- School of Pharmaceutical Sciences, São Paulo State University, Rod. Araraquara-Jaú, Araraquara, 14800903, Brazil
| | - Daniel A G Ruiz
- Institute of Chemistry, São Paulo State University, Rua Professor Francisco Degni, 55, Araraquara, 14800060, Brazil
| | - Nailton M Nascimento-Junior
- Institute of Chemistry, São Paulo State University, Rua Professor Francisco Degni, 55, Araraquara, 14800060, Brazil
| | - William A Denny
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Andrew M Thompson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Fernando R Pavan
- School of Pharmaceutical Sciences, São Paulo State University, Rod. Araraquara-Jaú, Araraquara, 14800903, Brazil
| | - Jean L Dos Santos
- School of Pharmaceutical Sciences, São Paulo State University, Rod. Araraquara-Jaú, Araraquara, 14800903, Brazil
| |
Collapse
|
12
|
Distel JS, Di Venanzio G, Mackel JJ, Rosen DA, Feldman MF. Replicative Acinetobacter baumannii strains interfere with phagosomal maturation by modulating the vacuolar pH. PLoS Pathog 2023; 19:e1011173. [PMID: 37294840 DOI: 10.1371/journal.ppat.1011173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/23/2023] [Indexed: 06/11/2023] Open
Abstract
Bacterial pneumonia is a common infection of the lower respiratory tract that can afflict patients of all ages. Multidrug-resistant strains of Acinetobacter baumannii are increasingly responsible for causing nosocomial pneumonias, thus posing an urgent threat. Alveolar macrophages play a critical role in overcoming respiratory infections caused by this pathogen. Recently, we and others have shown that new clinical isolates of A. baumannii, but not the common lab strain ATCC 19606 (19606), can persist and replicate in macrophages within spacious vacuoles that we called Acinetobacter Containing Vacuoles (ACV). In this work, we demonstrate that the modern A. baumannii clinical isolate 398, but not the lab strain 19606, can infect alveolar macrophages and produce ACVs in vivo in a murine pneumonia model. Both strains initially interact with the alveolar macrophage endocytic pathway, as indicated by EEA1 and LAMP1 markers; however, the fate of these strains diverges at a later stage. While 19606 is eliminated in an autophagy pathway, 398 replicates in ACVs and are not degraded. We show that 398 reverts the natural acidification of the phagosome by secreting large amounts of ammonia, a by-product of amino acid catabolism. We propose that this ability to survive within macrophages may be critical for the persistence of clinical A. baumannii isolates in the lung during a respiratory infection.
Collapse
Affiliation(s)
- Jesus S Distel
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Gisela Di Venanzio
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Joseph J Mackel
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - David A Rosen
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Mario F Feldman
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
13
|
D'Elia JA, Weinrauch LA. Gated Calcium Ion Channel and Mutation Mechanisms in Multidrug-Resistant Tuberculosis. Int J Mol Sci 2023; 24:ijms24119670. [PMID: 37298620 DOI: 10.3390/ijms24119670] [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: 02/28/2023] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
A wide spectrum of Gram-positive/Gram-negative bacteria has been found resistant to a wide spectrum of antibiotics in the United States of America during the past decade. Drug-resistant tuberculosis is not yet a major threat in North/South America, Europe, and the Middle East. However, the migration of populations in times of drought, famine, and hostilities may increase the global reach of this ancient pathogen. Given an increased spread from China and India to African countries, drug-resistant Mycobacterium tuberculosis has become an emerging topic of concern for Europe and North America. Due to the dangers associated with the spread of pathogens among different populations, the World Health Organization continues to expand healthcare advisories for therapeutic approaches for both stationary and migrating populations. As much of the literature focuses on endemic to pandemic viruses, we remain concerned that other treatable communicable diseases may be ignored. One such disease is multidrug-resistant tuberculosis. We focus on molecular mechanisms that this pathogen relies upon for the development of multidrug resistance via gene mutation and the evolutionary development of new enzyme and calcium channels.
Collapse
Affiliation(s)
- John A D'Elia
- Kidney/Hypertension Section, E P Joslin Research Laboratory, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Larry A Weinrauch
- Kidney/Hypertension Section, E P Joslin Research Laboratory, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
14
|
Carabalí-Isajar ML, Rodríguez-Bejarano OH, Amado T, Patarroyo MA, Izquierdo MA, Lutz JR, Ocampo M. Clinical manifestations and immune response to tuberculosis. World J Microbiol Biotechnol 2023; 39:206. [PMID: 37221438 DOI: 10.1007/s11274-023-03636-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/29/2023] [Indexed: 05/25/2023]
Abstract
Tuberculosis is a far-reaching, high-impact disease. It is among the top ten causes of death worldwide caused by a single infectious agent; 1.6 million tuberculosis-related deaths were reported in 2021 and it has been estimated that a third of the world's population are carriers of the tuberculosis bacillus but do not develop active disease. Several authors have attributed this to hosts' differential immune response in which cellular and humoral components are involved, along with cytokines and chemokines. Ascertaining the relationship between TB development's clinical manifestations and an immune response should increase understanding of tuberculosis pathophysiological and immunological mechanisms and correlating such material with protection against Mycobacterium tuberculosis. Tuberculosis continues to be a major public health problem globally. Mortality rates have not decreased significantly; rather, they are increasing. This review has thus been aimed at deepening knowledge regarding tuberculosis by examining published material related to an immune response against Mycobacterium tuberculosis, mycobacterial evasion mechanisms regarding such response and the relationship between pulmonary and extrapulmonary clinical manifestations induced by this bacterium which are related to inflammation associated with tuberculosis dissemination through different routes.
Collapse
Grants
- a Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia
- a Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia
- a Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia
- a Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia
- b PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Carrera 24#63C-69, Bogotá 111221, Colombia
- c Health Sciences Faculty, Universidad de Ciencias Aplicadas y Ambientales (UDCA), Calle 222#55-37, Bogotá 111166, Colombia
- d Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia
- e Medicine Department, Hospital Universitario Mayor Mederi, Calle 24 # 29-45, Bogotá 111411. Colombia
- e Medicine Department, Hospital Universitario Mayor Mederi, Calle 24 # 29-45, Bogotá 111411. Colombia
- f Universidad Distrital Francisco José de Caldas, Carrera 3#26A-40, Bogotá 110311, Colombia
Collapse
Affiliation(s)
- Mary Lilián Carabalí-Isajar
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, 111321, Bogotá, Colombia
- Biomedical and Biological Sciences Programme, Universidad del Rosario, Carrera 24#63C-69, 111221, Bogotá, Colombia
| | | | - Tatiana Amado
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, 111321, Bogotá, Colombia
| | - Manuel Alfonso Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, 111321, Bogotá, Colombia
- Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, 111321, Bogotá, Colombia
| | - María Alejandra Izquierdo
- Medicine Department, Hospital Universitario Mayor Mederi, Calle 24 # 29-45, 111411, Bogotá, Colombia
| | - Juan Ricardo Lutz
- Medicine Department, Hospital Universitario Mayor Mederi, Calle 24 # 29-45, 111411, Bogotá, Colombia.
| | - Marisol Ocampo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, 111321, Bogotá, Colombia.
- Universidad Distrital Francisco José de Caldas, Carrera 3#26A-40, 110311, Bogotá, Colombia.
| |
Collapse
|
15
|
Golovkine GR, Roberts AW, Morrison HM, Rivera-Lugo R, McCall RM, Nilsson H, Garelis NE, Repasy T, Cronce M, Budzik J, Van Dis E, Popov LM, Mitchell G, Zalpuri R, Jorgens D, Cox JS. Autophagy restricts Mycobacterium tuberculosis during acute infection in mice. Nat Microbiol 2023; 8:819-832. [PMID: 37037941 PMCID: PMC11027733 DOI: 10.1038/s41564-023-01354-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 03/03/2023] [Indexed: 04/12/2023]
Abstract
Whether or not autophagy has a role in defence against Mycobacterium tuberculosis infection remains unresolved. Previously, conditional knockdown of the core autophagy component ATG5 in myeloid cells was reported to confer extreme susceptibility to M. tuberculosis in mice, whereas depletion of other autophagy factors had no effect on infection. We show that doubling cre gene dosage to more robustly deplete ATG16L1 or ATG7 resulted in increased M. tuberculosis growth and host susceptibility in mice, although ATG5-depleted mice are more sensitive than ATG16L1- or ATG7-depleted mice. We imaged individual macrophages infected with M. tuberculosis and identified a shift from apoptosis to rapid necrosis in autophagy-depleted cells. This effect was dependent on phagosome permeabilization by M. tuberculosis. We monitored infected cells by electron microscopy, showing that autophagy protects the host macrophage by partially reducing mycobacterial access to the cytosol. We conclude that autophagy has an important role in defence against M. tuberculosis in mammals.
Collapse
Affiliation(s)
- Guillaume R Golovkine
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Evotec, Toulouse, France
| | - Allison W Roberts
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Huntly M Morrison
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Rafael Rivera-Lugo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Rita M McCall
- Department of Plant & Microbial Biology, University of California, Berkeley, CA, USA
| | - Hannah Nilsson
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Nicholas E Garelis
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Teresa Repasy
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Bio-Rad Laboratories, Seattle, WA, USA
| | - Michael Cronce
- Department of Bioengineering, University of California, Berkeley, CA, USA
- UC Berkeley-UCSF Graduate program in Bioengineering, Berkeley, CA, USA
| | - Jonathan Budzik
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Erik Van Dis
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Lauren M Popov
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Novome Biotechnologies, San Francisco, CA, USA
| | - Gabriel Mitchell
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Open Innovation @ NITD, Novartis Institute for Tropical Diseases, Emeryville, CA, USA
| | - Reena Zalpuri
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Danielle Jorgens
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Jeffery S Cox
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
16
|
Methylprednisolone Promotes Mycobacterium smegmatis Survival in Macrophages through NF-κB/DUSP1 Pathway. Microorganisms 2023; 11:microorganisms11030768. [PMID: 36985341 PMCID: PMC10058212 DOI: 10.3390/microorganisms11030768] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023] Open
Abstract
Background: Mycobacterium tuberculosis (M. tuberculosis) is the causative agent of tuberculosis. As an important component of host immunity, macrophages are not only the first line of defense against M. tuberculosis but also the parasitic site of M. tuberculosis in the host. Glucocorticoids can cause immunosuppression, which is considered to be one of the major risk factors for active tuberculosis, but the mechanism is unclear. Objective: To study the effect of methylprednisolone on the proliferation of mycobacteria in macrophages and try to find key molecules of this phenomenon. Methods: The macrophage line RAW264.7 infected by M. smegmatis was treated with methylprednisolone, and the intracellular bacterial CFU, Reactive Oxygen Species (ROS), cytokine secretion, autophagy, and apoptosis were measured. After the cells were treated with NF-κB inhibitor BAY 11-7082 and DUSP1 inhibitor BCI, respectively, the intracellular bacterial CFU, ROS, IL-6, and TNF-α secretion were detected. Results: After treatment with methylprednisolone, the CFU of intracellular bacteria increased, the level of ROS decreased, and the secretion of IL-6 and TNF-α decreased in infected macrophages. After BAY 11-7082 treatment, the CFU of M. smegmatis in macrophages increased, and the level of ROS production and the secretion of IL-6 by macrophages decreased. Transcriptome high-throughput sequencing and bioinformatics analysis suggested that DUSP1 was the key molecule in the above phenomenon. Western blot analysis confirmed that the expression level of DUSP1 was increased in the infected macrophages treated with methylprednisolone and BAY 11-7082, respectively. After BCI treatment, the level of ROS produced by infected macrophages increased, and the secretion of IL-6 increased. After the treatment of BCI combined with methylprednisolone or BAY 11-7082, the level of ROS produced and the secretion of IL-6 by macrophages were increased. Conclusion: methylprednisolone promotes the proliferation of mycobacteria in macrophages by suppressing cellular ROS production and IL-6 secretion through down-regulating NF-κB and up-regulating DUSP1 expression. BCI, an inhibitor of DUSP1, can reduce the level of DUSP1 in the infected macrophages and inhibit the proliferation of intracellular mycobacteria by promoting cellular ROS production and IL-6 secretion. Therefore, BCI may become a new molecule for host-directed therapy of tuberculosis, as well as a new strategy for the prevention of tuberculosis when treated with glucocorticoids.
Collapse
|
17
|
Wan H, Cai Y, Xiao L, Ling Y, Ge L, Mo S, Xie Q, Peng S, Zhou B, Zeng X, Chen X. JFD, a Novel Natural Inhibitor of Keap1 Alkylation, Suppresses Intracellular Mycobacterium Tuberculosis Growth through Keap1/Nrf2/SOD2-Mediated ROS Accumulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6726654. [PMID: 36819778 PMCID: PMC9937762 DOI: 10.1155/2023/6726654] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 08/16/2022] [Accepted: 10/14/2022] [Indexed: 02/12/2023]
Abstract
It is an effective strategy to treat tuberculosis by enhancing reactive oxygen species- (ROS-) mediated killing of Mycobacterium tuberculosis in macrophages, but there are no current therapeutic agents targeting this pathway. Honeysuckle has been used as the traditional medicine for tuberculosis treatment for 1500 years. Japoflavone D (JFD) is a novel biflavonoid isolated from Honeysuckle promoting ROS accumulation by Nrf2 pathway in hepatocarcinoma cells. However, its activity to kill M. tuberculosis in macrophages and molecular mechanism has not been reported. Our results showed that JFD enhances the M. tuberculosis elimination by boosting ROS levels in THP-1 cells. Moreover, the massive ROS accumulation activates p38 to induce apoptosis. Notably, the mechanism revealed that JFD suppresses the nuclear transport of Nrf2, thereby inhibiting SOD2 transcription, leading to a large ROS accumulation. Further studies showed that JFD disrupts the Keap1 alkylation at specific residues Cys14, Cys257, and Cys319, which is crucial for Nrf2 activation, thereby interrupts the nuclear transport of Nrf2. In pharmacokinetic study, JFD can stay as the prototype for 24 h in mice and can be excreted in feces without any toxicity. Our data reveal for the first time that a novel biflavonoid JFD as a potent inhibitor of Keap1 alkylation can suppress the nuclear transport of Nrf2. And it is the first research of the inhibitor of Keap1 alkylation. Furthermore, JFD robustly promotes M. tuberculosis elimination from macrophages by inhibiting Keap1/Nrf2/SOD2 pathway, resulting in the ROS accumulation. This work identified Keap1 alkylation as a new drug target for tuberculosis and provides a preliminary basis for the development of antituberculosis lead compounds based on JFD.
Collapse
Affiliation(s)
- Haoqiang Wan
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Lingyun Xiao
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Yunzhi Ling
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Lanlan Ge
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
| | - Siwei Mo
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Qiujie Xie
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Shusong Peng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Boping Zhou
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Xiaobin Zeng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| |
Collapse
|
18
|
Nanosized Drug Delivery Systems to Fight Tuberculosis. Pharmaceutics 2023; 15:pharmaceutics15020393. [PMID: 36839715 PMCID: PMC9964171 DOI: 10.3390/pharmaceutics15020393] [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: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Tuberculosis (TB) is currently the second deadliest infectious disease. Existing antitubercular therapies are long, complex, and have severe side effects that result in low patient compliance. In this context, nanosized drug delivery systems (DDSs) have the potential to optimize the treatment's efficiency while reducing its toxicity. Hundreds of publications illustrate the growing interest in this field. In this review, the main challenges related to the use of drug nanocarriers to fight TB are overviewed. Relevant publications regarding DDSs for the treatment of TB are classified according to the encapsulated drugs, from first-line to second-line drugs. The physicochemical and biological properties of the investigated formulations are listed. DDSs could simultaneously (i) optimize the therapy's antibacterial effects; (ii) reduce the doses; (iii) reduce the posology; (iv) diminish the toxicity; and as a global result, (v) mitigate the emergence of resistant strains. Moreover, we highlight that host-directed therapy using nanoparticles (NPs) is a recent promising trend. Although the research on nanosized DDSs for TB treatment is expanding, clinical applications have yet to be developed. Most studies are only dedicated to the development of new formulations, without the in vivo proof of concept. In the near future, it is expected that NPs prepared by "green" scalable methods, with intrinsic antibacterial properties and capable of co-encapsulating synergistic drugs, may find applications to fight TB.
Collapse
|
19
|
Ifijen IH, Atoe B, Ekun RO, Ighodaro A, Odiachi IJ. Treatments of Mycobacterium tuberculosis and Toxoplasma gondii with Selenium Nanoparticles. BIONANOSCIENCE 2023; 13:249-277. [PMID: 36687337 PMCID: PMC9838309 DOI: 10.1007/s12668-023-01059-4] [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] [Accepted: 01/02/2023] [Indexed: 01/13/2023]
Abstract
Toxoplasma gondii and Mycobacterium tuberculosis are pathogens that are harmful to humans. When these diseases interact in humans, the result is typically fatal to the public health. Several investigations on the relationship between M. tuberculosis and T. gondii infections have found that there is a strong correlation between them with each infection having a reciprocal effect on the other. TB may contribute to the reactivation of innate toxoplasmosis or enhance susceptibility to a new infection, and toxoplasma co-infection may worsen the severity of pulmonary tuberculosis. As a consequence, there is an earnest and urgent necessity to generate novel therapeutics that can subdue these challenges. Selenium nanostructures' compelling properties have been shown to be a successful treatment for Mycobacterium TB and Toxoplasma gondii. Despite the fact that selenium (Se) offers many health advantages for people, it also has a narrow therapeutic window; therefore, consuming too much of either inorganic or organic compounds based on selenium can be hazardous. Compared to both inorganic and organic Se, Se nanoparticles (SeNPs) are less hazardous. They are biocompatible and excellent in selectively targeting specific cells. As a consequence, this review conducted a summary of the efficacy of biogenic Se NPs in the treatment of tuberculosis (TB) and toxoplasmosis. Mycobacterium tuberculosis, Toxoplasma gondii, and their co-infection were all briefly described.
Collapse
Affiliation(s)
- Ikhazuagbe H. Ifijen
- Department of Research Outreach, Rubber Research Institute of Nigeria, Iyanomo, P.M.B, 1049, Benin City, Nigeria
| | - Best Atoe
- Department of Daily Need, Worldwide Healthcare, 100, Textile Mill Road, Benin City, Edo State Nigeria
| | - Raphael O. Ekun
- grid.440833.80000 0004 0642 9705Department of Electrical Electronics, Cyprus International University, Haspolat, Lefkosa, North Cyprus Mersin 10 Turkey
| | - Augustine Ighodaro
- Depatment of Aseptic Quality, Quantum Pharmaceuticals, Quantum House, Durham, UK
| | - Ifeanyi J. Odiachi
- grid.461933.a0000 0004 0446 5040Department of Science Laboratory Technology, Delta State Polytechnic Ogwashi-Uku, Ogwashi-Uku, Nigeria
| |
Collapse
|
20
|
Thakur M, Muniyappa K. Macrophage activation highlight an important role for NER proteins in the survival, latency and multiplication of Mycobacterium tuberculosis. Tuberculosis (Edinb) 2023; 138:102284. [PMID: 36459831 DOI: 10.1016/j.tube.2022.102284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/14/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
Nucleotide excision repair (NER) is one of the most extensively studied DNA repair processes in both prokaryotes and eukaryotes. The NER pathway is a highly conserved, ATP-dependent multi-step process involving several proteins/enzymes that function in a concerted manner to recognize and excise a wide spectrum of helix-distorting DNA lesions and bulky adducts by nuclease cleavage on either side of the damaged bases. As such, the NER pathway of Mycobacterium tuberculosis (Mtb) is essential for its survival within the hostile environment of macrophages and disease progression. This review focuses on present published knowledge about the crucial roles of Mtb NER proteins in the survival and multiplication of the pathogen within the macrophages and as potential targets for drug discovery.
Collapse
Affiliation(s)
- Manoj Thakur
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India.
| | - K Muniyappa
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
21
|
González K, Gangapurwala G, Alex J, Vollrath A, Cseresnyés Z, Weber C, Czaplewska JA, Hoeppener S, Svensson CM, Orasch T, Heinekamp T, Guerrero-Sánchez C, Figge MT, Schubert US, Brakhage AA. Targeting of phagolysosomes containing conidia of the fungus Aspergillus fumigatus with polymeric particles. Appl Microbiol Biotechnol 2023; 107:819-834. [PMID: 36480041 PMCID: PMC9842589 DOI: 10.1007/s00253-022-12287-1] [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: 08/02/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 12/13/2022]
Abstract
Conidia of the airborne human-pathogenic fungus Aspergillus fumigatus are inhaled by humans. In the lung, they are phagocytosed by alveolar macrophages and intracellularly processed. In macrophages, however, conidia can interfere with the maturation of phagolysosomes to avoid their elimination. To investigate whether polymeric particles (PPs) can reach this intracellular pathogen in macrophages, we formulated dye-labeled PPs with a size allowing for their phagocytosis. PPs were efficiently taken up by RAW 264.7 macrophages and were found in phagolysosomes. When macrophages were infected with conidia prior to the addition of PPs, we found that they co-localized in the same phagolysosomes. Mechanistically, the fusion of phagolysosomes containing PPs with phagolysosomes containing conidia was observed. Increasing concentrations of PPs increased fusion events, resulting in 14% of phagolysosomes containing both conidia and PPs. We demonstrate that PPs can reach conidia-containing phagolysosomes, making these particles a promising carrier system for antimicrobial drugs to target intracellular pathogens. KEY POINTS: • Polymer particles of a size larger than 500 nm are internalized by macrophages and localized in phagolysosomes. • These particles can be delivered to Aspergillus fumigatus conidia-containing phagolysosomes of macrophages. • Enhanced phagolysosome fusion by the use of vacuolin1 can increase particle delivery.
Collapse
Affiliation(s)
- Katherine González
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Adolf-Reichwein-Straße 23, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Neugasse 25, 07745 Jena, Germany
| | - Gauri Gangapurwala
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Julien Alex
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Antje Vollrath
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Zoltán Cseresnyés
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Adolf-Reichwein-Straße 23, 07745 Jena, Germany
| | - Christine Weber
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Justyna A. Czaplewska
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Carl-Magnus Svensson
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Adolf-Reichwein-Straße 23, 07745 Jena, Germany
| | - Thomas Orasch
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Adolf-Reichwein-Straße 23, 07745 Jena, Germany
| | - Thorsten Heinekamp
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Adolf-Reichwein-Straße 23, 07745 Jena, Germany
| | - Carlos Guerrero-Sánchez
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Marc Thilo Figge
- Institute of Microbiology, Friedrich Schiller University Jena, Neugasse 25, 07745 Jena, Germany
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Adolf-Reichwein-Straße 23, 07745 Jena, Germany
| | - Ulrich S. Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Axel A. Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Adolf-Reichwein-Straße 23, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Neugasse 25, 07745 Jena, Germany
| |
Collapse
|
22
|
Mouse Models for Mycobacterium tuberculosis Pathogenesis: Show and Do Not Tell. Pathogens 2022; 12:pathogens12010049. [PMID: 36678397 PMCID: PMC9865329 DOI: 10.3390/pathogens12010049] [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: 11/11/2022] [Revised: 11/29/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022] Open
Abstract
Science has been taking profit from animal models since the first translational experiments back in ancient Greece. From there, and across all history, several remarkable findings have been obtained using animal models. One of the most popular models, especially for research in infectious diseases, is the mouse. Regarding research in tuberculosis, the mouse has provided useful information about host and bacterial traits related to susceptibility to the infection. The effect of aging, sexual dimorphisms, the route of infection, genetic differences between mice lineages and unbalanced immunity scenarios upon Mycobacterium tuberculosis infection and tuberculosis development has helped, helps and will help biomedical researchers in the design of new tools for diagnosis, treatment and prevention of tuberculosis, despite various discrepancies and the lack of deep study in some areas of these traits.
Collapse
|
23
|
Jeong EK, Lee HJ, Jung YJ. Host-Directed Therapies for Tuberculosis. Pathogens 2022; 11:1291. [PMID: 36365041 PMCID: PMC9697779 DOI: 10.3390/pathogens11111291] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 02/04/2024] Open
Abstract
Tuberculosis (TB) is one of the leading causes of death worldwide, consistently threatening public health. Conventional tuberculosis treatment requires a long-term treatment regimen and is associated with side effects. The efficacy of antitubercular drugs has decreased with the emergence of drug-resistant TB; therefore, the development of new TB treatment strategies is urgently needed. In this context, we present host-directed therapy (HDT) as an alternative to current tuberculosis therapy. Unlike antitubercular drugs that directly target Mycobacterium tuberculosis (Mtb), the causative agent of TB, HDT is an approach for treating TB that appropriately modulates host immune responses. HDT primarily aims to enhance the antimicrobial activity of the host in order to control Mtb infection and attenuate excessive inflammation in order to minimize tissue damage. Recently, research based on the repositioning of drugs for use in HDT has been in progress. Based on the overall immune responses against Mtb infection and the immune-evasion mechanisms of Mtb, this review examines the repositioned drugs available for HDT and their mechanisms of action.
Collapse
Affiliation(s)
- Eui-Kwon Jeong
- BIT Medical Convergence Graduate Program, Kangwon National University, Chuncheon 24341, Korea
| | - Hyo-Ji Lee
- Department of Biological Sciences, Kangwon National University, Chuncheon 24341, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Yu-Jin Jung
- BIT Medical Convergence Graduate Program, Kangwon National University, Chuncheon 24341, Korea
- Department of Biological Sciences, Kangwon National University, Chuncheon 24341, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
24
|
Dong T, Chen X, Xu H, Song Y, Wang H, Gao Y, Wang J, Du R, Lou H, Dong T. Mitochondrial metabolism mediated macrophage polarization in chronic lung diseases. Pharmacol Ther 2022; 239:108208. [DOI: 10.1016/j.pharmthera.2022.108208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/01/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022]
|
25
|
Singh S, Maurya SK, Aqdas M, Bashir H, Arora A, Bhalla V, Agrewala JN. Mycobacterium tuberculosis exploits MPT64 to generate myeloid-derived suppressor cells to evade the immune system. Cell Mol Life Sci 2022; 79:567. [DOI: 10.1007/s00018-022-04596-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/24/2022]
|
26
|
Lin X, Jia W, Feng G, Su Y, Kang Y, Zhang C, Liu W, Lu Z, Xue D. The role of APTX4870 peptide in reducing cellular inflammatory responses by inhibiting Mycobacterium tuberculosis-derived mycolic acid-induced cytotoxicity. Front Microbiol 2022; 13:993897. [DOI: 10.3389/fmicb.2022.993897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis is a serious zoonotic disease caused by Mycobacterium tuberculosis (M.tb) and the M.tb complex. Mycolic acid is an extracellular carbohydrate polymer produced, secreted, and accumulated outside the cells of various Mycobacterium tuberculosis strains. Mycolic acid produced by Mycobacterium plays an important role in infection. However, there have been few reports on drugs that inhibit mycolic acid-induced cytotoxicity. The purpose of this study was to investigate the role of the panned peptide in Mycobacterium-derived mycolic acid (M.tb-MA)-induced cell injury. The heptapeptide (APTX4870) was isolated from various phage libraries using phage display (Ph.D-7, Ph.D-12, and Ph.D-C7C). The efficacy of APTX4870 against mycolic acid was demonstrated by evaluating clinical samples and conducting in vitro and Vivo. APTX4870 inhibited apoptosis, increased autophagy to decrease inflammation, and reduced M.tb-MA-induced lung damage. These findings suggest that this heptapeptide, which selectively targets M.tb-MA, might be exploited as a potential novel M.tb therapeutic treatment.
Collapse
|
27
|
Wang D, Lin Y, Xu F, Zhang H, Zhu X, Liu Z, Hu Y, Dong G, Sun B, Yu Y, Ma G, Tang Z, Legarda D, Ting A, Liu Y, Hou J, Dong L, Xiong H. SIRPα maintains macrophage homeostasis by interacting with PTK2B kinase in Mycobacterium tuberculosis infection and through autophagy and necroptosis. EBioMedicine 2022; 85:104278. [PMID: 36202053 PMCID: PMC9535427 DOI: 10.1016/j.ebiom.2022.104278] [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: 08/17/2021] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/11/2022] Open
Abstract
Background To determine whether SIRPα can be a diagnostic marker of pulmonary tuberculosis (PTB) and the molecular mechanism of SIRPα regulating macrophages to kill Mycobacterium tuberculosis (MTB). Methods Meta-analysis combined with subsequent qRT-PCR, western-blotting and flow cytometry assay were used to detect SIRPα expression in PTB patients. Cell-based assays were used to explore the regulation of macrophage function by SIRPα. SIRPα−/- and wide type macrophages transplanted C57BL/6J mice were used to determine the function of SIRPα on MTB infection in vivo. Findings SIRPα levels are closely correlated with the treatment outcomes among PTB patients. Cell-based assay demonstrated that MTB significantly induces the expression of SIRPα on macrophages. SIRPα deficiency enhances the killing ability of macrophages against MTB through processes that involve enhanced autophagy and reduced necroptosis of macrophages. Mechanistically, SIRPα forms a direct interaction with PTK2B through its intracellular C-terminal domain, thus inhibiting PTK2B activation in macrophages. Necroptosis inhibition due to SIRPα deficiency requires PTK2B activity. The transfer of SIRPα-deficient bone marrow-derived macrophages (BMDMs) into wild type mice resulted in a drop of bacterial load in the lungs but an enhancement of inflammatory lung damage, and the combination of ulinastatin and SIRPα−/−→WT treatment could decrease the inflammation and maintain the bactericidal capacity. Interpretation Our data define SIRPα a novel biomarker for tuberculosis infection and underlying mechanisms for maintaining macrophage homeostasis. Funding This work was financially supported by the Chinese National Natural Science Foundation project (No.81401635). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Collapse
Affiliation(s)
- Di Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, National Center for Liver Cancer, Shanghai, China,Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, America,The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yunkai Lin
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, National Center for Liver Cancer, Shanghai, China
| | - Feihong Xu
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, America
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining Shandong, China
| | - Xiaoyan Zhu
- The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhen Liu
- The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yuan Hu
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, America
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining Shandong, China
| | - Bingqi Sun
- Department of Clinical Laboratory, Shenyang Thoracic Hospital, Shenyang Liaoning, China
| | - Yanhong Yu
- Department of Clinical Laboratory, Shenyang Tenth People's Hospital, Shenyang Liaoning, China
| | - Guoren Ma
- Ningxia No. 4 People's Hospital, Yinchuan Ningxia, China
| | | | - Diana Legarda
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, America
| | - Adrian Ting
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, America
| | - Yuan Liu
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, America
| | - Jia Hou
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan Ningxia, China,Corresponding author at: Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan Ningxia, China.
| | - Liwei Dong
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, National Center for Liver Cancer, Shanghai, China,Corresponding author at: International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai, China.
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining Shandong, China,Corresponding author at: Institute of Immunology and Molecular Medicine, Jining Medical University, Jining Shandong, China.
| |
Collapse
|
28
|
Parbhoo T, Mouton JM, Sampson SL. Phenotypic adaptation of Mycobacterium tuberculosis to host-associated stressors that induce persister formation. Front Cell Infect Microbiol 2022; 12:956607. [PMID: 36237425 PMCID: PMC9551238 DOI: 10.3389/fcimb.2022.956607] [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: 05/30/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022] Open
Abstract
Mycobacterium tuberculosis exhibits a remarkable ability to interfere with the host antimicrobial response. The pathogen exploits elaborate strategies to cope with diverse host-induced stressors by modulating its metabolism and physiological state to prolong survival and promote persistence in host tissues. Elucidating the adaptive strategies that M. tuberculosis employs during infection to enhance persistence is crucial to understanding how varying physiological states may differentially drive disease progression for effective management of these populations. To improve our understanding of the phenotypic adaptation of M. tuberculosis, we review the adaptive strategies employed by M. tuberculosis to sense and coordinate a physiological response following exposure to various host-associated stressors. We further highlight the use of animal models that can be exploited to replicate and investigate different aspects of the human response to infection, to elucidate the impact of the host environment and bacterial adaptive strategies contributing to the recalcitrance of infection.
Collapse
|
29
|
Liebler-Tenorio EM, Heyl J, Wedlich N, Figl J, Köhler H, Krishnamoorthy G, Nieuwenhuizen NE, Grode L, Kaufmann SHE, Menge C. Vaccine-Induced Subcutaneous Granulomas in Goats Reflect Differences in Host-Mycobacterium Interactions between BCG- and Recombinant BCG-Derivative Vaccines. Int J Mol Sci 2022; 23:10992. [PMID: 36232295 PMCID: PMC9570401 DOI: 10.3390/ijms231910992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Tuberculous granulomas are highly dynamic structures reflecting the complex host-mycobacterium interactions. The objective of this study was to compare granuloma development at the site of vaccination with BCG and its recombinant derivatives in goats. To characterize the host response, epithelioid cells, multinucleated giant cells (MNGC), T cell subsets, B cells, plasma cells, dendritic cells and mycobacterial antigen were labelled by immunohistochemistry, and lipids and acid-fast bacteria (AFB) were labelled by specific staining. Granulomas with central caseous necrosis developed at the injection site of most goats though lesion size and extent of necrosis differed between vaccine strains. CD4+ T and B cells were more scarce and CD8+ cells were more numerous in granulomas induced by recombinant derivatives compared to their parental BCG strain. Further, the numbers of MNGCs and cells with lipid bodies were markedly lower in groups administered with recombinant BCG strains. Microscopic detection of AFB and mycobacterial antigen was rather frequent in the area of central necrosis, however, the isolation of bacteria in culture was rarely successful. In summary, BCG and its recombinant derivatives induced reproducibly subcutaneous caseous granulomas in goats that can be easily monitored and surgically removed for further studies. The granulomas reflected the genetic modifications of the recombinant BCG-derivatives and are therefore suitable models to compare reactions to different mycobacteria or TB vaccines.
Collapse
Affiliation(s)
| | - Johannes Heyl
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, 07743 Jena, Germany
| | - Nadine Wedlich
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, 07743 Jena, Germany
| | - Julia Figl
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, 07743 Jena, Germany
| | - Heike Köhler
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, 07743 Jena, Germany
| | | | | | - Leander Grode
- Vakzine Projekt Management GmbH, 30625 Hannover, Germany
| | - Stefan H. E. Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX 77843, USA
| | - Christian Menge
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, 07743 Jena, Germany
| |
Collapse
|
30
|
Singh VK, Chau E, Mishra A, DeAnda A, Hegde VL, Sastry JK, Haviland D, Jagannath C, Godin B, Khan A. CD44 receptor targeted nanoparticles augment immunity against tuberculosis in mice. J Control Release 2022; 349:796-811. [PMID: 35914613 PMCID: PMC10478167 DOI: 10.1016/j.jconrel.2022.07.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023]
Abstract
We describe a role of CD44-mediated signaling during host-defense against tuberculosis (TB) using a mouse model of TB and studies in M. tuberculosis (Mtb) infected human macrophage (MФ). Liposomes targeting CD44 using thioaptamers (CD44TA-LIP) were designed and tested as new vaccines to boost host immunity in TB. CD44TA-LIP enhanced killing of Mtb in human MФ, which correlated with an increased production of pro-inflammatory cytokines IL-1β, TNF-α and IL-12. CD44TA-LIP activated MФ showed an enhanced MHC-II dependent antigen presentation to CD4 T-cells. Inhibition of cellular proliferation and cytoskeleton rearrangement pathways downstream of CD44 signaling abrogated CD44TA-LIP-induced antimicrobial effects. Blockade of inflammatory pathways also reduced antigen presentation by MФ and activation of CD4 T cells. Mtb infected MФ treated with CD44TA-LIP exhibited increased nitric oxide and HβD2 defensin peptide production. Among Mtb infected mice with increased lung and spleen loads of organisms, intranasal administration of CD44TA-LIP led to a ten-fold reduction of colony forming units of Mtb and elevated IFN-γ + CD4, effector, central and resident memory T cells. Biodistribution studies demonstrated that CD44TA-LIP preferentially accumulated in the lungs and were associated with CD11b + cells. CD44TA-LIP treated mice showed no weight loss or increased liver LDH levels. This study highlights the importance of CD44-mediated signaling in host-defense during TB and the therapeutic potential of CD44TA-LIP.
Collapse
Affiliation(s)
- Vipul K Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Eric Chau
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Alexandro DeAnda
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Venkatesh L Hegde
- Department of Thoracic Head & Neck Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Jagannadha K Sastry
- Department of Thoracic Head & Neck Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - David Haviland
- Flow Cytometry Core, Houston Methodist Research Institute, Houston, TX, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA.
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
31
|
Human pluripotent stem cell-derived macrophages host Mycobacterium abscessus infection. Stem Cell Reports 2022; 17:2156-2166. [PMID: 35985333 PMCID: PMC9481898 DOI: 10.1016/j.stemcr.2022.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/22/2022] Open
Abstract
Human macrophages are a natural host of many mycobacterium species, including Mycobacterium abscessus (M. abscessus), an emerging pathogen affecting immunocompromised and cystic fibrosis patients with few available treatments. The search for an effective treatment is hindered by the lack of a tractable in vitro intracellular infection model. Here, we established a reliable model for M. abscessus infection using human pluripotent stem cell-derived macrophages (hPSC-macrophages). hPSC differentiation permitted reproducible generation of functional macrophages that were highly susceptible to M. abscessus infection. Electron microscopy demonstrated that M. abscessus was present in the hPSC-macrophage vacuoles. RNA sequencing analysis revealed a time-dependent host cell response, with differing gene and protein expression patterns post-infection. Engineered tdTOMATO-expressing hPSC-macrophages with GFP-expressing mycobacteria enabled rapid image-based high-throughput analysis of intracellular infection and quantitative assessment of antibiotic efficacy. Our study describes the first to our knowledge hPSC-based model for M. abscessus infection, representing a novel and accessible system for studying pathogen-host interaction and drug discovery. A simplified chemically defined and serum-free protocol for the generation of functional macrophages from hPSCs An efficient human model recapitulating intracellular infection of Mycobacterium abscessus in hPSC-macrophages A high-throughput system testing antibiotic sensitivity with fluorescent hPSC-macrophages and M. abscessus
Collapse
|
32
|
Paulowski L, Beckham KSH, Johansen MD, Berneking L, Van N, Degefu Y, Staack S, Sotomayor FV, Asar L, Rohde H, Aldridge BB, Aepfelbacher M, Parret A, Wilmanns M, Kremer L, Combrink K, Maurer FP. C25-modified rifamycin derivatives with improved activity against Mycobacterium abscessus. PNAS NEXUS 2022; 1:pgac130. [PMID: 36714853 PMCID: PMC9802118 DOI: 10.1093/pnasnexus/pgac130] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/05/2022] [Indexed: 02/01/2023]
Abstract
Infections caused by Mycobacterium abscessus are difficult to treat due to its intrinsic resistance to most antibiotics. Formation of biofilms and the capacity of M. abscessus to survive inside host phagocytes further complicate eradication. Herein, we explored whether addition of a carbamate-linked group at the C25 position of rifamycin SV blocks enzymatic inactivation by ArrMab, an ADP-ribosyltransferase conferring resistance to rifampicin (RMP). Unlike RMP, 5j, a benzyl piperidine rifamycin derivative with a morpholino substituted C3 position and a naphthoquinone core, is not modified by purified ArrMab. Additionally, we show that the ArrMab D82 residue is essential for catalytic activity. Thermal profiling of ArrMab in the presence of 5j, RMP, or rifabutin shows that 5j does not bind to ArrMab. We found that the activity of 5j is comparable to amikacin against M. abscessus planktonic cultures and pellicles. Critically, 5j also exerts potent antimicrobial activity against M. abscessus in human macrophages and shows synergistic activity with amikacin and azithromycin.
Collapse
Affiliation(s)
| | | | | | | | - Nhi Van
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine and Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Boston, MA 02111, USA
| | - Yonatan Degefu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine and Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Boston, MA 02111, USA
| | - Sonja Staack
- European Molecular Biology Laboratory, 22607 Hamburg, Germany
| | - Flor Vasquez Sotomayor
- National and WHO Supranational Reference Center for Mycobacteria, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany,Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lucia Asar
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Holger Rohde
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Bree B Aldridge
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine and Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Boston, MA 02111, USA
| | - Martin Aepfelbacher
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Annabel Parret
- European Molecular Biology Laboratory, 22607 Hamburg, Germany,Charles River Laboratories, 2340 Beerse, Belgium
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, 22607 Hamburg, Germany,University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 34293 Montpellier, France,INSERM, Institut de Recherche en Infectiologie de Montpellier, 34293 Montpellier, France
| | - Keith Combrink
- Department of Chemistry and Biochemistry, Texas A&M International University, Laredo, TX 77843, USA,Department of Chemistry, Blinn College, Bryan Campus, Brenham, TX 77833, USA
| | | |
Collapse
|
33
|
Bar-Oz M, Meir M, Barkan D. Virulence-Associated Secretion in Mycobacterium abscessus. Front Immunol 2022; 13:938895. [PMID: 35880173 PMCID: PMC9308005 DOI: 10.3389/fimmu.2022.938895] [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: 05/08/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Non-tuberculous mycobacteria (NTM) are a heterogeneous group of originally environmental organi3sms, increasingly recognized as pathogens with rising prevalence worldwide. Knowledge of NTM’s mechanisms of virulence is lacking, as molecular research of these bacteria is challenging, sometimes more than that of M. tuberculosis (Mtb), and far less resources are allocated to their investigation. While some of the virulence mechanisms are common to several mycobacteria including Mtb, others NTM species-specific. Among NTMs, Mycobacterium abscessus (Mabs) causes some of the most severe and difficult to treat infections, especially chronic pulmonary infections. Mabs survives and proliferates intracellularly by circumventing host defenses, using multiple mechanisms, many of which remain poorly characterized. Some of these immune-evasion mechanisms are also found in Mtb, including phagosome pore formation, inhibition of phagosome maturation, cytokine response interference and apoptosis delay. While much is known of the role of Mtb-secreted effector molecules in mediating the manipulation of the host response, far less is known of the secreted effector molecules in Mabs. In this review, we briefly summarize the knowledge of secreted effectors in Mtb (such as ESX secretion, SecA2, TAT and others), and draw the parallel pathways in Mabs. We also describe pathways that are unique to Mabs, differentiating it from Mtb. This review will assist researchers interested in virulence-associated secretion in Mabs by providing the knowledge base and framework for their studies.
Collapse
Affiliation(s)
- Michal Bar-Oz
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Michal Meir
- The Ruth Rappaport Children’s Hospital, Rambam Medical Center, Haifa, Israel
| | - Daniel Barkan
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- *Correspondence: Daniel Barkan,
| |
Collapse
|
34
|
Trousil J, Dal NJK, Fenaroli F, Schlachet I, Kubíčková P, Janoušková O, Pavlova E, Škorič M, Trejbalová K, Pavliš O, Sosnik A. Antibiotic-Loaded Amphiphilic Chitosan Nanoparticles Target Macrophages and Kill an Intracellular Pathogen. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201853. [PMID: 35691939 DOI: 10.1002/smll.202201853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/19/2022] [Indexed: 06/15/2023]
Abstract
In this work, levofloxacin (LVX), a third-generation fluoroquinolone antibiotic, is encapsulated within amphiphilic polymeric nanoparticles of a chitosan-g-poly(methyl methacrylate) produced by self-assembly and physically stabilized by ionotropic crosslinking with sodium tripolyphosphate. Non-crosslinked nanoparticles display a size of 29 nm and a zeta-potential of +36 mV, while the crosslinked counterparts display 45 nm and +24 mV, respectively. The cell compatibility, uptake, and intracellular trafficking are characterized in the murine alveolar macrophage cell line MH-S and the human bronchial epithelial cell line BEAS-2B in vitro. Internalization events are detected after 10 min and the uptake is inhibited by several endocytosis inhibitors, indicating the involvement of complex endocytic pathways. In addition, the nanoparticles are detected in the lysosomal compartment. Then, the antibacterial efficacy of LVX-loaded nanoformulations (50% w/w drug content) is assessed in MH-S and BEAS-2B cells infected with Staphylococcus aureus and the bacterial burden is decreased by 49% and 46%, respectively. In contrast, free LVX leads to a decrease of 8% and 5%, respectively, in the same infected cell lines. Finally, intravenous injection to a zebrafish larval model shows that the nanoparticles accumulate in macrophages and endothelium and demonstrate the promise of these amphiphilic nanoparticles to target intracellular infections.
Collapse
Affiliation(s)
- Jiří Trousil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, 162 00, Czech Republic
| | | | | | - Inbar Schlachet
- Laboratory of Pharmaceutical Nanomaterials Science, Faculty of Materials Science and Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Pavla Kubíčková
- Military Health Institute, Military Medical Agency, Prague, 160 00, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, 162 00, Czech Republic
- Department of Biology, Faculty of Science, University of J. E. Purkyně, Ústí nad Labem, 400 96, Czech Republic
| | - Ewa Pavlova
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, 162 00, Czech Republic
| | - Miša Škorič
- Department of Pathological Morphology and Parasitology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Brno, 612 42, Czech Republic
| | - Kateřina Trejbalová
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague, 142 20, Czech Republic
| | - Oto Pavliš
- Military Health Institute, Military Medical Agency, Prague, 160 00, Czech Republic
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Faculty of Materials Science and Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| |
Collapse
|
35
|
Azlyna ASN, Ahmad S, Husna SMN, Sarmiento ME, Acosta A, Norazmi MN, Mohamud R, Kadir R. Review: Liposomes in the prophylaxis and treatment of infectious diseases. Life Sci 2022; 305:120734. [PMID: 35760094 DOI: 10.1016/j.lfs.2022.120734] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 06/08/2022] [Accepted: 06/22/2022] [Indexed: 11/15/2022]
Abstract
Infectious diseases remain as one of the major burdens among health communities as well as in the general public despite the advances in prevention and treatment. Although vaccination and vector eliminations have greatly prevented the transmission of these diseases, the effectiveness of these strategies is no longer guaranteed as new challenges such as drug resistance and toxicity as well as the missing effective therapeutics arise. Hence, the development of new tools to manage these challenges is anticipated, in which nano technology using liposomes as effective nanostructure is highly considered. In this review, we concentrate on the advantages of liposomes in the drug delivery system and the development of vaccine in the treatment of three major infectious diseases; tuberculosis (TB), malaria and HIV.
Collapse
Affiliation(s)
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Siti Muhamad Nur Husna
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Maria E Sarmiento
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Armando Acosta
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Mohd Nor Norazmi
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
36
|
Magoulopoulou A, Qian X, Pediatama Setiabudiawan T, Marco Salas S, Yokota C, Rottenberg ME, Nilsson M, Carow B. Spatial Resolution of Mycobacterium tuberculosis Bacteria and Their Surrounding Immune Environments Based on Selected Key Transcripts in Mouse Lungs. Front Immunol 2022; 13:876321. [PMID: 35663950 PMCID: PMC9157500 DOI: 10.3389/fimmu.2022.876321] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) bacilli are the causative agent of tuberculosis (TB), a major killer of mankind. Although it is widely accepted that local interactions between Mtb and the immune system in the tuberculous granuloma determine whether the outcome of infection is controlled or disseminated, these have been poorly studied due to methodological constraints. We have recently used a spatial transcriptomic technique, in situ sequencing (ISS), to define the spatial distribution of immune transcripts in TB mouse lungs. To further contribute to the understanding of the immune microenvironments of Mtb and their local diversity, we here present two complementary automated bacteria-guided analysis pipelines. These position 33 ISS-identified immune transcripts in relation to single bacteria and bacteria clusters. The analysis was applied on new ISS data from lung sections of Mtb-infected C57BL/6 and C3HeB/FeJ mice. In lungs from C57BL/6 mice early and late post infection, transcripts that define inflammatory macrophages were enriched at subcellular distances to bacteria, indicating the activation of infected macrophages. In contrast, expression patterns associated to antigen presentation were enriched in non-infected cells at 12 weeks post infection. T-cell transcripts were evenly distributed in the tissue. In Mtb-infected C3HeB/FeJ mice, transcripts characterizing activated macrophages localized in apposition to small bacteria clusters, but not in organized granulomas. Despite differences in the susceptibility to Mtb, the transcript patterns found around small bacteria clusters of C3HeB/FeJ and C57BL/6 mice were similar. Altogether, the presented tools allow us to characterize in depth the immune cell populations and their activation that interact with Mtb in the infected lung.
Collapse
Affiliation(s)
- Anastasia Magoulopoulou
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Xiaoyan Qian
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Todia Pediatama Setiabudiawan
- Department of Microbiology, Tumor and Cell Biology and Centre for Tuberculosis Research, Karolinska Institutet, Solna, Sweden
| | - Sergio Marco Salas
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Chika Yokota
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology and Centre for Tuberculosis Research, Karolinska Institutet, Solna, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Berit Carow
- Department of Microbiology, Tumor and Cell Biology and Centre for Tuberculosis Research, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
37
|
Khoza LJ, Kumar P, Dube A, Demana PH, Choonara YE. Insights into Innovative Therapeutics for Drug-Resistant Tuberculosis: Host-Directed Therapy and Autophagy Inducing Modified Nanoparticles. Int J Pharm 2022; 622:121893. [PMID: 35680110 PMCID: PMC9169426 DOI: 10.1016/j.ijpharm.2022.121893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 10/25/2022]
|
38
|
Dwivedi M, Bajpai K. The chamber of secretome in Mycobacterium tuberculosis as a potential therapeutic target. Biotechnol Genet Eng Rev 2022; 39:1-44. [PMID: 35613080 DOI: 10.1080/02648725.2022.2076031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mycobacterium tuberculosis (MTB) causes one of the ancient diseases, Tuberculosis, affects people around the globe and its severity can be understood by its classification as a second infectious disease after COVID-19 and the 13th leading cause of death according to a WHO report. Despite having advanced diagnostic approaches and therapeutic strategies, unfortunately, TB is still spreading across the population due to the emergence of drug-resistance MTB and Latent TB infection (LTBI). We are seeking for effective approaches to overcome these hindrances and efficient treatment for this perilous disease. Therefore, there is an urgent need to develop drugs based on operative targeting of the bacterial system that could result in both efficient treatment and lesser emergence of MDR-TB. One such promising target could be the secretory systems and especially the Type 7 secretory system (T7SS-ESX) of Mycobacterium tuberculosis, which is crucial for the secretion of effector proteins as well as in establishing host-pathogen interactions of the tubercle bacilli. The five paralogous ESX systems (ESX-1 to EXS-5) have been observed by in silico genome analysis of MTB, among which ESX-1 and ESX-5 are substantial for virulence and mediating host cellular inflammasome. The bacterium growth and virulence can be modulated by targeting the T7SS. In the present review, we demonstrate the current status of therapeutics against MTB and focus on the function and cruciality of T7SS along with other secretory systems as a promising therapeutic target against Tuberculosis.
Collapse
Affiliation(s)
- Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Kriti Bajpai
- Department of Biotechnology, Himachal Pradesh University, Shimla, India
| |
Collapse
|
39
|
Fernandes GFS, Thompson AM, Castagnolo D, Denny WA, Dos Santos JL. Tuberculosis Drug Discovery: Challenges and New Horizons. J Med Chem 2022; 65:7489-7531. [PMID: 35612311 DOI: 10.1021/acs.jmedchem.2c00227] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Over the past 2000 years, tuberculosis (TB) has claimed more lives than any other infectious disease. In 2020 alone, TB was responsible for 1.5 million deaths worldwide, comparable to the 1.8 million deaths caused by COVID-19. The World Health Organization has stated that new TB drugs must be developed to end this pandemic. After decades of neglect in this field, a renaissance era of TB drug discovery has arrived, in which many novel candidates have entered clinical trials. However, while hundreds of molecules are reported annually as promising anti-TB agents, very few successfully progress to clinical development. In this Perspective, we critically review those anti-TB compounds published in the last 6 years that demonstrate good in vivo efficacy against Mycobacterium tuberculosis. Additionally, we highlight the main challenges and strategies for developing new TB drugs and the current global pipeline of drug candidates in clinical studies to foment fresh research perspectives.
Collapse
Affiliation(s)
- Guilherme F S Fernandes
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - Andrew M Thompson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Daniele Castagnolo
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - William A Denny
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jean L Dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800903, Brazil
| |
Collapse
|
40
|
Matteucci KC, Correa AAS, Costa DL. Recent Advances in Host-Directed Therapies for Tuberculosis and Malaria. Front Cell Infect Microbiol 2022; 12:905278. [PMID: 35669122 PMCID: PMC9163498 DOI: 10.3389/fcimb.2022.905278] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/21/2022] [Indexed: 11/30/2022] Open
Abstract
Tuberculosis (TB), caused by the bacterium Mycobacterium tuberculosis, and malaria, caused by parasites from the Plasmodium genus, are two of the major causes of death due to infectious diseases in the world. Both diseases are treatable with drugs that have microbicidal properties against each of the etiologic agents. However, problems related to treatment compliance by patients and emergence of drug resistant microorganisms have been a major problem for combating TB and malaria. This factor is further complicated by the absence of highly effective vaccines that can prevent the infection with either M. tuberculosis or Plasmodium. However, certain host biological processes have been found to play a role in the promotion of infection or in the pathogenesis of each disease. These processes can be targeted by host-directed therapies (HDTs), which can be administered in conjunction with the standard drug treatments for each pathogen, aiming to accelerate their elimination or to minimize detrimental side effects resulting from exacerbated inflammation. In this review we discuss potential new targets for the development of HDTs revealed by recent advances in the knowledge of host-pathogen interaction biology, and present an overview of strategies that have been tested in vivo, either in experimental models or in patients.
Collapse
Affiliation(s)
- Kely C. Matteucci
- Plataforma de Medicina Translacional Fundação Oswaldo Cruz/Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - André A. S. Correa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Diego L. Costa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Diego L. Costa,
| |
Collapse
|
41
|
Ahmad F, Rani A, Alam A, Zarin S, Pandey S, Singh H, Hasnain SE, Ehtesham NZ. Macrophage: A Cell With Many Faces and Functions in Tuberculosis. Front Immunol 2022; 13:747799. [PMID: 35603185 PMCID: PMC9122124 DOI: 10.3389/fimmu.2022.747799] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/30/2022] [Indexed: 01/16/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of human tuberculosis (TB) which primarily infects the macrophages. Nearly a quarter of the world's population is infected latently by Mtb. Only around 5%-10% of those infected develop active TB disease, particularly during suppressed host immune conditions or comorbidity such as HIV, hinting toward the heterogeneity of Mtb infection. The aerosolized Mtb first reaches the lungs, and the resident alveolar macrophages (AMs) are among the first cells to encounter the Mtb infection. Evidence suggests that early clearance of Mtb infection is associated with robust innate immune responses in resident macrophages. In addition to lung-resident macrophage subsets, the recruited monocytes and monocyte-derived macrophages (MDMs) have been suggested to have a protective role during Mtb infection. Mtb, by virtue of its unique cell surface lipids and secreted protein effectors, can evade killing by the innate immune cells and preferentially establish a niche within the AMs. Continuous efforts to delineate the determinants of host defense mechanisms have brought to the center stage the crucial role of macrophage phenotypical variations for functional adaptations in TB. The morphological and functional heterogeneity and plasticity of the macrophages aid in confining the dissemination of Mtb. However, during a suppressed or hyperactivated immune state, the Mtb virulence factors can affect macrophage homeostasis which may skew to favor pathogen growth, causing active TB. This mini-review is aimed at summarizing the interplay of Mtb pathomechanisms in the macrophages and the implications of macrophage heterogeneity and plasticity during Mtb infection.
Collapse
Affiliation(s)
- Faraz Ahmad
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Anshu Rani
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
| | - Anwar Alam
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Sheeba Zarin
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Saurabh Pandey
- Department of Biochemistry, Jamia Hamdard, New Delhi, India
| | - Hina Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, India
| | - Nasreen Zafar Ehtesham
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| |
Collapse
|
42
|
Jurkowitz MS, Azad AK, Monsma PC, Keiser TL, Kanyo J, Lam TT, Bell CE, Schlesinger LS. Mycobacterium tuberculosis encodes a YhhN family membrane protein with lysoplasmalogenase activity that protects against toxic host lysolipids. J Biol Chem 2022; 298:101849. [PMID: 35314194 PMCID: PMC9052158 DOI: 10.1016/j.jbc.2022.101849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/09/2022] Open
Abstract
The pathogen Mycobacterium tuberculosis (M.tb) resides in human macrophages, wherein it exploits host lipids for survival. However, little is known about the interaction between M.tb and macrophage plasmalogens, a subclass of glycerophospholipids with a vinyl ether bond at the sn-1 position of the glycerol backbone. Lysoplasmalogens, produced from plasmalogens by hydrolysis at the sn-2 carbon by phospholipase A2, are potentially toxic but can be broken down by host lysoplasmalogenase, an integral membrane protein of the YhhN family that hydrolyzes the vinyl ether bond to release a fatty aldehyde and glycerophospho-ethanolamine or glycerophospho-choline. Curiously, M.tb encodes its own YhhN protein (MtbYhhN), despite having no endogenous plasmalogens. To understand the purpose of this protein, the gene for MtbYhhN (Rv1401) was cloned and expressed in Mycobacterium smegmatis (M.smeg). We found the partially purified protein exhibited abundant lysoplasmalogenase activity specific for lysoplasmenylethanolamine or lysoplasmenylcholine (pLPC) (Vmax∼15.5 μmol/min/mg; Km∼83 μM). Based on cell density, we determined that lysoplasmenylethanolamine, pLPC, lysophosphatidylcholine, and lysophosphatidylethanolamine were not toxic to M.smeg cells, but pLPC and LPC were highly toxic to M.smeg spheroplasts, which are cell wall-deficient mycobacterial forms. Importantly, spheroplasts prepared from M.smeg cells overexpressing MtbYhhN were protected from membrane disruption/lysis by pLPC, which was rapidly depleted from the media. Finally, we found that overexpression of full-length MtbYhhN in M.smeg increased its survival within human macrophages by 2.6-fold compared to vector controls. These data support the hypothesis that MtbYhhN protein confers a growth advantage for mycobacteria in macrophages by cleaving toxic host pLPC into potentially energy-producing products.
Collapse
Affiliation(s)
- Marianne S Jurkowitz
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, Ohio, USA.
| | - Abul K Azad
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Paula C Monsma
- Department of Neuroscience, Ohio State University, Columbus, Ohio, USA
| | - Tracy L Keiser
- Department of Moleculaire Microbiologie, Vrije Universiteit, Amsterdam, the Netherlands
| | - Jean Kanyo
- Keck MS & Proteomics Resource, Yale University, New Haven, Connecticut, USA
| | - TuKiet T Lam
- Keck MS & Proteomics Resource, Yale University, New Haven, Connecticut, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Charles E Bell
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, Ohio, USA; Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Larry S Schlesinger
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, Texas, USA.
| |
Collapse
|
43
|
RNA Microarray-Based Comparison of Innate Immune Phenotypes between Human THP-1 Macrophages Stimulated with Two BCG Strains. Int J Mol Sci 2022; 23:ijms23094525. [PMID: 35562916 PMCID: PMC9103163 DOI: 10.3390/ijms23094525] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 12/10/2022] Open
Abstract
Currently, the only available vaccine against tuberculosis is Mycobacterium bovis Bacille Calmette-Guérin (BCG). Pulmonary tuberculosis protection provided by the vaccine varies depending on the strain, the patient’s age and the evaluated population. Although the adaptive immune responses induced by different BCG strains have been widely studied, little conclusive data is available regarding innate immune responses, especially in macrophages. Here, we aimed to characterize the innate immune responses of human THP-1-derived macrophages at the transcriptional level following a challenge with either the BCG Mexico (M.BCG) or Phipps (P.BCG) strains. After a brief in vitro characterization of the bacterial strains and the innate immune responses, including nitric oxide production and cytokine profiles, we analyzed the mRNA expression patterns and performed pathway enrichment analysis using RNA microarrays. Our results showed that multiple biological processes were enriched, especially those associated with innate inflammatory and antimicrobial responses, including tumor necrosis factor (TNF)-α, type I interferon (IFN-I) and IFN-γ. However, four DEGs were identified in macrophages infected with M.BCG compared to P. BCG. These findings indicated the proinflammatory stimulation of macrophages induced by both BCG strains, at the cytokine level and in terms of gene expression, suggesting a differential expression pattern of innate immune transcripts depending on the mycobacterial strain.
Collapse
|
44
|
Niesteruk A, Sreeramulu S, Jonker HRA, Richter C, Schwalbe H. Oxidation of the Mycobacterium tuberculosis key virulence factor Protein Tyrosine Phosphatase A (MptpA) reduces its phosphatase activity. FEBS Lett 2022; 596:1503-1515. [PMID: 35397176 DOI: 10.1002/1873-3468.14348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/06/2022]
Abstract
The Mycobacterium tuberculosis tyrosine-specific phosphatase MptpA and its cognate kinase PtkA are prospective targets for anti- tuberculosis drugs as they interact with the host defense response within the macrophages. Although both are structurally well characterized, the functional mechanism regulating their activity remains poorly understood. Here, we investigate the effect of post-translational oxidation in regulating the function of MptpA. Treatment of MptpA with H2 O2 /NaHCO3 , mimicking cellular oxidative stress conditions, leads to oxidation of the catalytic cysteine (C11) and to a conformational rearrangement of the phosphorylation loop (D-loop) by repositioning the conserved tyrosine 128 (Y128) and generating a temporarily inactive pre-closed state of the phosphatase. Thus, the catalytic cysteine in the P-loop acts as a redox switch and regulates the phosphatase activity of MptpA.
Collapse
Affiliation(s)
- Anna Niesteruk
- Goethe University Frankfurt am Main, Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Frankfurt am Main, Germany
| | - Sridhar Sreeramulu
- Goethe University Frankfurt am Main, Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Frankfurt am Main, Germany
| | - Hendrik R A Jonker
- Goethe University Frankfurt am Main, Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Frankfurt am Main, Germany
| | - Christian Richter
- Goethe University Frankfurt am Main, Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Frankfurt am Main, Germany
| | - Harald Schwalbe
- Goethe University Frankfurt am Main, Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Frankfurt am Main, Germany
| |
Collapse
|
45
|
Pharmacoengineered Lipid Core–Shell Nanoarchitectonics to Influence Human Alveolar Macrophages Uptake for Drug Targeting Against Tuberculosis. J Inorg Organomet Polym Mater 2022. [DOI: 10.1007/s10904-022-02306-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
46
|
Host cell targeting of novel antimycobacterial 4-aminosalicylic acid derivatives with tuftsin carrier peptides. Eur J Pharm Biopharm 2022; 174:111-130. [DOI: 10.1016/j.ejpb.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/08/2022] [Accepted: 03/24/2022] [Indexed: 11/23/2022]
|
47
|
Herb M, Gluschko A, Farid A, Krönke M. When the Phagosome Gets Leaky: Pore-Forming Toxin-Induced Non-Canonical Autophagy (PINCA). Front Cell Infect Microbiol 2022; 12:834321. [PMID: 35372127 PMCID: PMC8968195 DOI: 10.3389/fcimb.2022.834321] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Macrophages remove bacteria from the extracellular milieu via phagocytosis. While most of the engulfed bacteria are degraded in the antimicrobial environment of the phagolysosome, several bacterial pathogens have evolved virulence factors, which evade degradation or allow escape into the cytosol. To counter this situation, macrophages activate LC3-associated phagocytosis (LAP), a highly bactericidal non-canonical autophagy pathway, which destroys the bacterial pathogens in so called LAPosomes. Moreover, macrophages can also target intracellular bacteria by pore-forming toxin-induced non-canonical autophagy (PINCA), a recently described non-canonical autophagy pathway, which is activated by phagosomal damage induced by bacteria-derived pore-forming toxins. Similar to LAP, PINCA involves LC3 recruitment to the bacteria-containing phagosome independently of the ULK complex, but in contrast to LAP, this process does not require ROS production by Nox2. As last resort of autophagic targeting, macrophages activate xenophagy, a selective form of macroautophagy, to recapture bacteria, which evaded successful targeting by LAP or PINCA through rupture of the phagosome. However, xenophagy can also be hijacked by bacterial pathogens for their benefit or can be completely inhibited resulting in intracellular growth of the bacterial pathogen. In this perspective, we discuss the molecular differences and similarities between LAP, PINCA and xenophagy in macrophages during bacterial infections.
Collapse
Affiliation(s)
- Marc Herb
- Faculty of Medicine and University Hospital of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Alexander Gluschko
- Faculty of Medicine and University Hospital of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Alina Farid
- Faculty of Medicine and University Hospital of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Martin Krönke
- Faculty of Medicine and University Hospital of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- German Center for Infection Research, Bonn-Cologne, Germany
| |
Collapse
|
48
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
49
|
Hunter L, Hingley-Wilson S, Stewart GR, Sharpe SA, Salguero FJ. Dynamics of Macrophage, T and B Cell Infiltration Within Pulmonary Granulomas Induced by Mycobacterium tuberculosis in Two Non-Human Primate Models of Aerosol Infection. Front Immunol 2022; 12:776913. [PMID: 35069548 PMCID: PMC8770544 DOI: 10.3389/fimmu.2021.776913] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/14/2021] [Indexed: 12/25/2022] Open
Abstract
Non-human primate models of Tuberculosis (TB) are one of the most commonly used within the experimental TB field because they closely mimic the whole spectrum of disease progression of human TB. However, the early cellular interactions of the pulmonary granuloma are still not well understood. The use of this model allows investigation into the early interactions of cells within pulmonary granulomas which cannot be undertaken in human samples. Pulmonary granulomas from rhesus and cynomolgus macaques from two timepoints post infection were categorised into categories 1 – 6 (early to late stage granulomas) and immunohistochemistry was used to identify CD68+ macrophages, CD3+ T cells and CD20+ B cells. Multinucleated giant cells and acid-fast bacilli were also quantified. At week four post infection, cynomolgus macaques were found to have more CD68+ cells than rhesus in all but category 1 granulomas. Cynomolgus also had a significantly higher percentage of CD20+ B cells in category 1 granulomas. At week twelve post infection, CD68+ cells were most abundant in category 4 and 5 granulomas in both species; however, there were no significant differences between them. CD3+ T cells and CD20+ B cells were significantly higher in the majority of granuloma categories in cynomolgus compared to rhesus. Multinucleated giant cells and acid-fast bacilli were most abundant in categories 5 and 6 at week 12 post challenge in both species. This study has identified the basic cellular composition and spatial distribution of immune cells within pulmonary granulomas in both rhesus and cynomolgus macaques over time. The data from this study will add to the knowledge already gained in this field and may inform future research on vaccines and therapeutics for TB.
Collapse
Affiliation(s)
- Laura Hunter
- Research and Evaluation, UK Health Security Agency (UKHSA), Salisbury, United Kingdom.,School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Suzie Hingley-Wilson
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Graham R Stewart
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Sally A Sharpe
- Research and Evaluation, UK Health Security Agency (UKHSA), Salisbury, United Kingdom
| | | |
Collapse
|
50
|
Petit TJ, Lebreton A. Adaptations of intracellular bacteria to vacuolar or cytosolic niches. Trends Microbiol 2022; 30:736-748. [DOI: 10.1016/j.tim.2022.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/28/2022]
|