1
|
Bromley JD, Ganchua SKC, Nyquist SK, Maiello P, Chao M, Borish HJ, Rodgers M, Tomko J, Kracinovsky K, Mugahid D, Nguyen S, Wang QD, Rosenberg JM, Klein EC, Gideon HP, Floyd-O'Sullivan R, Berger B, Scanga CA, Lin PL, Fortune SM, Shalek AK, Flynn JL. CD4 + T cells re-wire granuloma cellularity and regulatory networks to promote immunomodulation following Mtb reinfection. Immunity 2024; 57:2380-2398.e6. [PMID: 39214090 PMCID: PMC11466276 DOI: 10.1016/j.immuni.2024.08.002] [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: 12/22/2023] [Revised: 06/03/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Immunological priming-in the context of either prior infection or vaccination-elicits protective responses against subsequent Mycobacterium tuberculosis (Mtb) infection. However, the changes that occur in the lung cellular milieu post-primary Mtb infection and their contributions to protection upon reinfection remain poorly understood. Using clinical and microbiological endpoints in a non-human primate reinfection model, we demonstrated that prior Mtb infection elicited a long-lasting protective response against subsequent Mtb exposure and was CD4+ T cell dependent. By analyzing data from primary infection, reinfection, and reinfection-CD4+ T cell-depleted granulomas, we found that the presence of CD4+ T cells during reinfection resulted in a less inflammatory lung milieu characterized by reprogrammed CD8+ T cells, reduced neutrophilia, and blunted type 1 immune signaling among myeloid cells. These results open avenues for developing vaccines and therapeutics that not only target lymphocytes but also modulate innate immune cells to limit tuberculosis (TB) disease.
Collapse
Affiliation(s)
- Joshua D Bromley
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sharie Keanne C Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah K Nyquist
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Chao
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - H Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jaime Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kara Kracinovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Douaa Mugahid
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Son Nguyen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Qianchang Dennis Wang
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob M Rosenberg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Edwin C Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hannah P Gideon
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Roisin Floyd-O'Sullivan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Philana Ling Lin
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah M Fortune
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Patel A, Nguyen L, Shea C, Singh S, Venketaraman V. The Role of mTOR in Mycobacterium tuberculosis Infection. Biomedicines 2024; 12:2238. [PMID: 39457551 PMCID: PMC11505195 DOI: 10.3390/biomedicines12102238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Mycobacterium tuberculosis (M. tb) is a pathogen that causes tuberculosis (TB), an extremely infectious disease which is responsible for millions of deaths worldwide. The severity of this pathogen is further amplified with the emergence of multidrug-resistant strains that are becoming more prevalent at an alarming rate, and novel treatments are needed. Methods: In this paper, we discuss the pathology M. tb infection. We review the literature on the role that mTOR plays in autophagy and the immune system as well as its impact on M. tb infection. Lastly, we discuss the current therapies targeting mTOR and potential routes to explore for future treatments. Results: The mTOR protein acts as a negative regulator of the autophagy pathway and presents as a potent target to establish new treatments for TB. M. tb survival is affected by mTOR, the PI3K/mTOR/AKT pathway, and autophagy. M. tb evades destruction by manipulating host cellular mechanisms, which increases resistance and complicates treatment. Conclusions: Targeting mTOR can enhance autophagy and increase M. tb clearance. Existing drugs such as everolimus, rapamycin + CC214-2, and bazedoxifene are all being currently studied for effectiveness and show positive results. Alternative therapies, including Chinese herbs, baicalin, BTLA, glutathione, and precision medicine can modulate the PI3K/mTOR/AKT pathway and the host's immune response, resulting in increased M. tb clearance, and these may be the future treatments for M. tb infection.
Collapse
Affiliation(s)
| | | | | | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.P.); (L.N.); (C.S.); (S.S.)
| |
Collapse
|
3
|
Branchett WJ, Saraiva M, O'Garra A. Regulation of inflammation by Interleukin-10 in the intestinal and respiratory mucosa. Curr Opin Immunol 2024; 91:102495. [PMID: 39357078 DOI: 10.1016/j.coi.2024.102495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
Intricate immune regulation is required at mucosal surfaces to allow tolerance to microbiota and harmless allergens and to prevent overexuberant inflammatory responses to pathogens. The cytokine Interleukin-10 (IL-10) is a key mediator of mucosal immune regulation. While IL-10 can be produced by virtually all cells of the immune system, many of its in vivo functions depend upon its production by regulatory or effector T cell populations and its signalling to macrophages, dendritic cells and specific T cell subsets. In this review, we discuss our current understanding of the role of IL-10 in regulation of immune responses, with a focus on its context-specific roles in intestinal homeostasis, respiratory infection and asthma. We highlight the importance of appropriate production and function of IL-10 for balancing pathogen clearance, control of microbiota and host tissue damage, and that precise modulation of IL-10 functions in vivo could present therapeutic opportunities.
Collapse
Affiliation(s)
- William J Branchett
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, United Kingdom.
| | - Margarida Saraiva
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Anne O'Garra
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
4
|
Seo H, Yoon Y, Kim S, Ghorbanian F, Tajdozian H, Jo S, Barman I, Lee S, Lee Y, Rahim MA, Hossain MS, Lee S, Song HY. Anti-tuberculosis effect of microbiome therapeutic PMC205 in extensively drug-resistant pulmonary tuberculosis in vivo. Int J Antimicrob Agents 2024; 64:107274. [PMID: 39002701 DOI: 10.1016/j.ijantimicag.2024.107274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 06/05/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Tuberculosis is a highly contagious disease caused by Mycobacterium tuberculosis, and the increase in antibiotic resistance threatens humankind. Therefore, there is an urgent need to develop new anti-tuberculosis drugs that can overcome the limitations of existing drugs. Here, we report the anti-tuberculosis effect of microbiome therapeutic PMC205, a strain of Bacillus subtilis. METHODS The anti-tuberculosis activity of probiotics was evaluated in mouse models of lethal and latent pulmonary tuberculosis induced by high or low-dose infection of the extensively drug-resistant strain. Probiotics were administered by inhalation, and the burden of M. tuberculosis in the lungs, along with mortality and clinical observations, were monitored for 12 weeks and 8 months, respectively. For an in-depth understanding, analysis of the microbiome and inflammatory profile of the lung microenvironment and induction of autophagy in vitro were explored. RESULTS After inhalation administration of PMC205 for 3 months, the survival rate was 100%, unlike all deaths in the saline-treated group, and the burden of M. tuberculosis in the lungs was reduced by log 1.3 in the 8-month latent tuberculosis model. Moreover, PMC205 induced recovery of disrupted lung microflora, increased butyric acid, and suppressed excessive inflammation. It also promoted autophagy. CONCLUSIONS These results confirm PMC205's anti-tuberculosis effect, suggesting that it can be developed as an adjuvant to current antibiotic therapy to solve the drug-resistant tuberculosis problem.
Collapse
Affiliation(s)
- Hoonhee Seo
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Youjin Yoon
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Sukyung Kim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Fatemeh Ghorbanian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Hanieh Tajdozian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Sujin Jo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Soyeon Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Youngkyoung Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Saebim Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea; Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea; Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea.
| |
Collapse
|
5
|
Chen Z, Zhang Y, Wu J, Xu J, Hu Z, Fan XY. A multistage protein subunit vaccine as BCG-booster confers protection against Mycobacterium tuberculosis infection in murine models. Int Immunopharmacol 2024; 139:112811. [PMID: 39068754 DOI: 10.1016/j.intimp.2024.112811] [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: 04/21/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
The eradication of tuberculosis remains a global challenge. Despite being the only licensed vaccine, Bacillus Calmette-Guérin (BCG) confers limited protective efficacy in adults and individuals with latent tuberculosis infections (LTBI). There is an urgent need to develop novel vaccines that can enhance the protective effect of BCG. Protein subunit vaccines have garnered significant research interest due to their safety and plasticity. Based on previous studies, we selected three antigens associated with LTBI (Rv2028c, Rv2029c, Rv3126c) and fused them with an immunodominant antigen Ag85A, resulting in the construction of a multistage protein subunit vaccine named A986. We evaluated the protective effect of recombinant protein A986 adjuvanted with MPL/QS21 as a booster vaccine for BCG against Mycobacterium tuberculosis (Mtb) infection in mice. The A986 + MPL/QS21 induced the secretion of antigen-specific Th1 (IL-2+, IFN-γ+ and TNF-α+) and Th17 (IL-17A+) cytokines in CD4+ and CD8+ T cells within the lung and spleen of mice, while also increased the frequency of central memory and effector memory T cells. Additionally, it also induced the enhanced production of IgG antibodies. Compared to BCG alone, A986 + MPL/QS21 boosting significantly augmented the proliferation of antigen-specific multifunctional T cells and effectively reduced bacterial load in infected mice. Taken together, A986 + MPL/QS21 formulation induced robust antigen-specific immune responses and provided enhanced protection against Mtb infection as a booster of BCG vaccine.
Collapse
Affiliation(s)
- Zhenyan Chen
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital & The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Ying Zhang
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Juan Wu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Jinchuan Xu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Zhidong Hu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China.
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital & The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China.
| |
Collapse
|
6
|
Piccaro G, Aquino G, Gigantino V, Tirelli V, Sanchez M, Iorio E, Matarese G, Cassone A, Palma C. Mycobacterium tuberculosis antigen 85B modifies BCG-induced antituberculosis immunity and favors pathogen survival. J Leukoc Biol 2024; 115:1053-1069. [PMID: 38242866 DOI: 10.1093/jleuko/qiae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Tuberculosis is one of the deadliest infectious diseases worldwide. Mycobacterium tuberculosis has developed strategies not only to evade host immunity but also to manipulate it for its survival. We investigated whether Mycobacterium tuberculosis exploited the immunogenicity of Ag85B, one of its major secretory proteins, to redirect host antituberculosis immunity to its advantage. We found that administration of Ag85B protein to mice vaccinated with Bacillus Calmette-Guérin impaired the protection elicited by vaccination, causing a more severe infection when mice were challenged with Mycobacterium tuberculosis. Ag85B administration reduced Bacillus Calmette-Guérin-induced CD4 T-cell activation and IFN-γ, CCL-4, and IL-22 production in response to Mycobacterium tuberculosis-infected cells. On the other hand, it promoted robust Ag85B-responsive IFN-γ-producing CD4 T cells, expansion of a subset of IFN-γ/IL-10-producing CD4+FOXP3+Treg cells, differential activation of IL-17/IL-22 responses, and activation of regulatory and exhaustion pathways, including programmed death ligand 1 expression on macrophages. All this resulted in impaired intracellular Mycobacterium tuberculosis growth control by systemic immunity, both before and after the Mycobacterium tuberculosis challenge. Interestingly, Mycobacterium tuberculosis infection itself generated Ag85B-reactive inflammatory immune cells incapable of clearing Mycobacterium tuberculosis in both unvaccinated and Bacillus Calmette-Guérin-vaccinated mice. Our data suggest that Mycobacterium tuberculosis can exploit the strong immunogenicity of Ag85B to promote its own survival and spread. Since Ag85B is normally secreted by replicating bacteria and is commonly found in the lungs of the Mycobacterium tuberculosis-infected host, our findings may advance the understanding on the mechanisms of Mycobacterium tuberculosis pathogenesis and immune evasion.
Collapse
Affiliation(s)
- Giovanni Piccaro
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Gabriella Aquino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Valentina Tirelli
- Core Facilities-Flow Cytometry Area, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Massimo Sanchez
- Core Facilities-Flow Cytometry Area, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Egidio Iorio
- Core Facilities-High Resolution NMR Unit, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie mediche, Università di Napoli "Federico II," Via Sergio Pansini 5, 80131 Naples, Italy
| | - Antonio Cassone
- Polo d'innovazione della Genomica, Genetica e Biologia, Via Fiorentina 1, 53100 Siena, Italy
| | - Carla Palma
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
7
|
Bahlool AZ, Cavanagh B, Sullivan AO, MacLoughlin R, Keane J, Sullivan MPO, Cryan SA. Microfluidics produced ATRA-loaded PLGA NPs reduced tuberculosis burden in alveolar epithelial cells and enabled high delivered dose under simulated human breathing pattern in 3D printed head models. Eur J Pharm Sci 2024; 196:106734. [PMID: 38417586 DOI: 10.1016/j.ejps.2024.106734] [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: 09/26/2023] [Revised: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/01/2024]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is second only to COVID-19 as the top infectious disease killer worldwide. Multi-drug resistant TB (MDR-TB) may arise because of poor patient adherence to medications due to lengthy treatment duration and side effects. Delivering novel host directed therapies (HDT), like all trans retinoic acid (ATRA) may help to improve drug regimens and reduce the incidence of MDR-TB. Local delivery of ATRA to the site of infection leads to higher bioavailability and reduced systemic side effects. ATRA is poorly soluble in water and has a short half-life in plasma. Therefore, it requires a formulation step before it can be administered in vivo. ATRA loaded PLGA nanoparticles suitable for nebulization were manufactured and optimized using a scalable nanomanufacturing microfluidics (MF) mixing approach (MF-ATRA-PLGA NPs). MF-ATRA-PLGA NPs demonstrated a dose dependent inhibition of Mtb growth in TB-infected A549 alveolar epithelial cell model while preserving cell viability. The MF-ATRA-PLGA NPs were nebulized with the Aerogen Solo vibrating mesh nebulizer, with aerosol droplet size characterized using laser diffraction and the estimated delivered dose was determined. The volume median diameter (VMD) of the MF-ATRA-PLGA NPs was 3.00 ± 0.18 μm. The inhaled dose delivered in adult and paediatric 3D printed head models under a simulated normal adult and paediatric breathing pattern was found to be 47.05 ± 3 % and 20.15 ± 3.46 % respectively. These aerosol characteristics of MF-ATRA-PLGA NPs supports its suitability for delivery to the lungs via inhalation. The data generated on the efficacy of an inhalable, scalable and regulatory friendly ATRA-PLGA NPs formulation provides a foundation on which further pre-clinical testing can be built. Overall, the results of this project are promising for future research into ATRA loaded NPs formulations as inhaled host directed therapies for TB.
Collapse
Affiliation(s)
- Ahmad Z Bahlool
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin, Ireland; Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland RCSI, Dublin 2, Ireland
| | - Andrew O' Sullivan
- Research and Development, Science and Emerging Technologies, Aerogen Ltd, Galway Business Park, Dangan, Galway, Ireland
| | - Ronan MacLoughlin
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Research and Development, Science and Emerging Technologies, Aerogen Ltd, Galway Business Park, Dangan, Galway, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Mary P O' Sullivan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin, Ireland; SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI and Trinity College Dublin, Dublin, Ireland; SFI Centre for Research in Medical Devices (CÚRAM), NUIG & RCSI, Dublin, Ireland.
| |
Collapse
|
8
|
Ullah H, Shi X, Taj A, Cheng L, Yan Q, Sha S, Ahmad, Kang J, Haris M, Ma X, Ma Y. Mycobacterium tuberculosis PE_PGRS38 Enhances Intracellular Survival of Mycobacteria by Inhibiting TLR4/NF-κB-Dependent Inflammation and Apoptosis of the Host. BIOLOGY 2024; 13:313. [PMID: 38785795 PMCID: PMC11118070 DOI: 10.3390/biology13050313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/20/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
Mycobacterium tuberculosis (Mtb) ranks as the most lethal human pathogen, able to fend off repeated attacks by the immune system or medications. PE_PGRS proteins are hallmarks of the pathogenicity of Mtb and contribute to its antigenic diversity, virulence, and persistence during infection. M. smegmatis is a nonpathogenic mycobacterium that naturally lacks PE_PGRS and is used as a model to express Mtb proteins. PE_PGRS has the capability to evade host immune responses and enhance the intracellular survival of M. smegmatis. Despite the intense investigations into PE_PGRS proteins, their role in tuberculosis remains elusive. We engineered the recombinant M. smegmatis strain Ms-PE_PGRS38. The result shows that PE_PGRS38 is expressed in the cell wall of M. smegmatis. PE_PGRS38 contributes to biofilm formation, confers permeability to the cell wall, and shows variable responses to exogenous stresses. PE_PGRS38 downregulated TLR4/NF-κB signaling in RAW264.7 macrophages and lung tissues of infected mice. In addition, PE_PGRS38 decreased NLRP3-dependent IL-1β release and limited pathogen-mediated inflammasome activity during infection. Moreover, PE_PGRS38 inhibited the apoptosis of RAW264.7 cells by downregulating the expression of apoptotic markers including Bax, cytochrome c, caspase-3, and caspase-9. In a nutshell, our findings demonstrate that PE_PGRS38 is a virulence factor for Mtb that enables recombinant M. smegmatis to survive by resisting and evading the host's immune responses during infection.
Collapse
Affiliation(s)
- Hayan Ullah
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
- Department of Microbiology, Dalian Medical University, Dalian 116044, China;
| | - Xiaoxia Shi
- Department of Experimental Teaching Center of Public Health, Dalian Medical University, Dalian 116044, China;
| | - Ayaz Taj
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Lin Cheng
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Qiulong Yan
- Department of Microbiology, Dalian Medical University, Dalian 116044, China;
| | - Shanshan Sha
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Ahmad
- Department of Immunology, Dalian Medical University, Dalian 116044, China;
| | - Jian Kang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Muhammad Haris
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Xiaochi Ma
- Pharmaceutical Research Center, The Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Yufang Ma
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
- Department of Microbiology, Dalian Medical University, Dalian 116044, China;
| |
Collapse
|
9
|
Yan Q, Zhao W, Kong H, Chi J, Dai Z, Yu D, Cui J. CT‑based radiomics analysis of consolidation characteristics in differentiating pulmonary disease of non‑tuberculous mycobacterium from pulmonary tuberculosis. Exp Ther Med 2024; 27:112. [PMID: 38361522 PMCID: PMC10867735 DOI: 10.3892/etm.2024.12400] [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: 06/28/2023] [Accepted: 11/02/2023] [Indexed: 02/17/2024] Open
Abstract
Global incidence rate of non-tuberculous mycobacteria (NTM) pulmonary disease has been increasing rapidly. In some countries and regions, its incidence rate is higher than that of tuberculosis. It is easily confused with tuberculosis. The topic of this study is to identify two diseases using CT radioomics. The aim in the present study was to investigate the value of CT-based radiomics to analyze consolidation features in differentiation of non-tuberculous mycobacteria (NTM) from pulmonary tuberculosis (TB). A total of 156 patients (75 with NTM pulmonary disease and 81 with TB) exhibiting consolidation characteristics in Shandong Public Health Clinical Center were retrospectively analyzed. Subsequently, 305 regions of interest of CT consolidation were outlined. Using a random number generated via a computer, 70 and 30% of consolidations were allocated to the training and the validation cohort, respectively. By means of variance threshold, when investigating the effective radiomics features, SelectKBest and the least absolute shrinkage and selection operator regression method were employed for feature selection and combined to calculate the radiomics score. K-nearest neighbor (KNN), support vector machine (SVM) and logistic regression (LR) were used to analyze effective radiomics features. A total of 18 patients with NTM pulmonary disease and 18 with TB possessing consolidation characteristics in Jinan Infectious Disease Hospital were collected for external validation of the model. A total of three methods was used in the selection of 52 optimal features. For KNN, the area under the curve (AUC; sensitivity, specificity) for the training and validation cohorts were 0.98 (0.93, 0.94) and 0.90 (0.88, 083), respectively; for SVM, AUC was 0.99 (0.96, 0.96) and 0.92 (0.86, 0.85) and for LR, AUC was 0.99 (0.97, 0.97) and 0.89 (0.88, 0.85). In the external validation cohort, AUC values of models were all >0.84 and LR classifier exhibited the most significant precision, recall and F1 score (0.87, 0.94 and 0.88, respectively). LR classifier possessed the best performance in differentiating diseases. Therefore, CT-based radiomics analysis of consolidation features may distinguish NTM pulmonary disease from TB.
Collapse
Affiliation(s)
- Qinghu Yan
- Department of Radiology, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Wenlong Zhao
- Department of Radiology, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Haili Kong
- Department of Radiology, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Jingyu Chi
- Department of Radiology, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Zhengjun Dai
- Huiying Medical Technology (Beijing) Co., Ltd., Beijing 100192, P.R. China
| | - Dexin Yu
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jia Cui
- Department of Radiology, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
10
|
Ruelas Castillo J, Neupane P, Karanika S, Krug S, Quijada D, Garcia A, Ayeh S, Yilma A, Costa DL, Sher A, Fotouhi N, Serbina N, Karakousis PC. The heme oxygenase-1 metalloporphyrin inhibitor stannsoporfin enhances the bactericidal activity of a novel regimen for multidrug-resistant tuberculosis in a murine model. Antimicrob Agents Chemother 2024; 68:e0104323. [PMID: 38132181 PMCID: PMC10848751 DOI: 10.1128/aac.01043-23] [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: 08/10/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
Multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) poses significant challenges to global tuberculosis (TB) control efforts. Host-directed therapies (HDTs) offer a novel approach to TB treatment by enhancing immune-mediated clearance of Mtb. Prior preclinical studies found that the inhibition of heme oxygenase-1 (HO-1), an enzyme involved in heme metabolism, with tin-protoporphyrin IX (SnPP) significantly reduced mouse lung bacillary burden when co-administered with the first-line antitubercular regimen. Here, we evaluated the adjunctive HDT activity of a novel HO-1 inhibitor, stannsoporfin (SnMP), in combination with a novel MDR-TB regimen comprising a next-generation diarylquinoline, TBAJ-876 (S), pretomanid (Pa), and a new oxazolidinone, TBI-223 (O) (collectively, SPaO), in Mtb-infected BALB/c mice. After 4 weeks of treatment, SPaO + SnMP 5mg/kg reduced mean lung bacillary burden by an additional 0.69 log10 (P = 0.01) relative to SPaO alone. As early as 2 weeks post-treatment initiation, SnMP adjunctive therapy differentially altered the expression of pro-inflammatory cytokine genes and CD38, a marker of M1 macrophages. Next, we evaluated the sterilizing potential of SnMP adjunctive therapy in a mouse model of microbiological relapse. After 6 weeks of treatment, SPaO + SnMP 10mg/kg reduced lung bacterial burdens to 0.71 ± 0.23 log10 colony-forming units (CFUs), a 0.78 log-fold greater decrease in lung CFU compared to SpaO alone (P = 0.005). However, adjunctive SnMP did not reduce microbiological relapse rates after 5 or 6 weeks of treatment. SnMP was well tolerated and did not significantly alter gross or histological lung pathology. SnMP is a promising HDT candidate requiring further study in combination with regimens for drug-resistant TB.
Collapse
Affiliation(s)
- Jennie Ruelas Castillo
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pranita Neupane
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Styliani Karanika
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stefanie Krug
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Darla Quijada
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Garcia
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samuel Ayeh
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Addis Yilma
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Diego L. Costa
- Departmento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Petros C. Karakousis
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Shi C, Huang X, Wang D, Chu C, Shi Y, Yan B, Shan F, Zhang J, Zhang Z, Peng C, Tang BZ. Lipophilic AIEgens as the "Trojan Horse" with Discrepant Efficacy in Tracking and Treatment of Mycobacterial Infection. Adv Healthc Mater 2024; 13:e2301746. [PMID: 37747232 DOI: 10.1002/adhm.202301746] [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: 06/01/2023] [Revised: 08/07/2023] [Indexed: 09/26/2023]
Abstract
The highly contagious tuberculosis is a leading infectious killer, which urgently requires effective diagnosis and treatment methods. To address these issues, three lipophilic aggregation-induced emission (AIE) photosensitizers (TTMN, TTTMN, and MeOTTMN) are selected to evaluate their labeling and antimicrobial properties in vitro and in vivo. These three lipophilic AIEgens preserve low cytotoxicity and achieve real-time and non-invasive visualization of the process of mycobacteria infection in vitro and in vivo. More importantly, these AIEgens can be triggered by white light to produce reactive oxygen species (ROS), which is a highly efficient antibacterial reagent. Among these AIEgens, the TTMN photosensitizer has an outstanding antibacterial efficacy over the clinical first-line drug rifampicin at the same therapeutic concentration. Interestingly, this study also finds that TTMN can increase the expression of pro-inflammatory cytokines in the early stage of infection after light irradiation, indicating an additional pro-inflammatory role of TTMN. This work provides some feasibility basis for developing AIEgens-based agents for effectively destroying mycobacterium.
Collapse
Affiliation(s)
- Chunzi Shi
- Qingdao Institute, School of Life Medicine, Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Qingdao, 266500, China
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Xueni Huang
- Qingdao Institute, School of Life Medicine, Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Qingdao, 266500, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Chengshengze Chu
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Yuxin Shi
- Qingdao Institute, School of Life Medicine, Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Qingdao, 266500, China
| | - Bo Yan
- Qingdao Institute, School of Life Medicine, Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Qingdao, 266500, China
| | - Fei Shan
- Qingdao Institute, School of Life Medicine, Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Qingdao, 266500, China
| | - Jiulong Zhang
- Qingdao Institute, School of Life Medicine, Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Qingdao, 266500, China
| | - Zhiyong Zhang
- Qingdao Institute, School of Life Medicine, Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Qingdao, 266500, China
| | - Chen Peng
- Qingdao Institute, School of Life Medicine, Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Qingdao, 266500, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, 518172, China
| |
Collapse
|
12
|
Bromley JD, Ganchua SKC, Nyquist SK, Maiello P, Chao M, Borish HJ, Rodgers M, Tomko J, Kracinovsky K, Mugahid D, Nguyen S, Wang D, Rosenberg JM, Klein EC, Gideon HP, Floyd-O’Sullivan R, Berger B, Scanga CA, Lin PL, Fortune SM, Shalek AK, Flynn JL. CD4 + T cells are homeostatic regulators during Mtb reinfection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572669. [PMID: 38187598 PMCID: PMC10769325 DOI: 10.1101/2023.12.20.572669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Immunological priming - either in the context of prior infection or vaccination - elicits protective responses against subsequent Mycobacterium tuberculosis (Mtb) infection. However, the changes that occur in the lung cellular milieu post-primary Mtb infection and their contributions to protection upon reinfection remain poorly understood. Here, using clinical and microbiological endpoints in a non-human primate reinfection model, we demonstrate that prior Mtb infection elicits a long-lasting protective response against subsequent Mtb exposure and that the depletion of CD4+ T cells prior to Mtb rechallenge significantly abrogates this protection. Leveraging microbiologic, PET-CT, flow cytometric, and single-cell RNA-seq data from primary infection, reinfection, and reinfection-CD4+ T cell depleted granulomas, we identify differential cellular and microbial features of control. The data collectively demonstrate that the presence of CD4+ T cells in the setting of reinfection results in a reduced inflammatory lung milieu characterized by reprogrammed CD8+ T cell activity, reduced neutrophilia, and blunted type-1 immune signaling among myeloid cells, mitigating Mtb disease severity. These results open avenues for developing vaccines and therapeutics that not only target CD4+ and CD8+ T cells, but also modulate innate immune cells to limit Mtb disease.
Collapse
Affiliation(s)
- Joshua D. Bromley
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sharie Keanne C. Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Sarah K. Nyquist
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
| | - Michael Chao
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - H. Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Mark Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Jaime Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Kara Kracinovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Douaa Mugahid
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Son Nguyen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dennis Wang
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob M. Rosenberg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Edwin C. Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hannah P. Gideon
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Roisin Floyd-O’Sullivan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Philana Ling Lin
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine
| | - Sarah M. Fortune
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alex K. Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
- Lead contact
| |
Collapse
|
13
|
Castillo JR, Neupane P, Karanika S, Krug S, Quijada D, Garcia A, Ayeh S, Yilma A, Costa DL, Sher A, Fotouhi N, Serbina N, Karakousis PC. The heme oxygenase-1 metalloporphyrin inhibitor stannsoporfin enhances the bactericidal activity of a novel regimen for multidrug-resistant tuberculosis in a murine model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552716. [PMID: 37609351 PMCID: PMC10441415 DOI: 10.1101/2023.08.09.552716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) poses significant challenges to global tuberculosis (TB) control efforts. Host-directed therapies (HDT) offer a novel approach for TB treatment by enhancing immune-mediated clearance of Mtb. Prior preclinical studies found that inhibition of heme oxygenase-1 (HO-1), an enzyme involved in heme metabolism, with tin-protoporphyrin IX (SnPP) significantly reduced mouse lung bacillary burden when co-administered with the first-line antitubercular regimen. Here we evaluated the adjunctive HDT activity of a novel HO-1 inhibitor, stannsoporfin (SnMP), in combination with a novel MDR-TB regimen comprising a next-generation diarylquinoline, TBAJ-876 (S), pretomanid (Pa), and a new oxazolidinone, TBI-223 (O) (collectively, SPaO) in Mtb-infected BALB/c mice. After 4 weeks of treatment, SPaO + SnMP 5 mg/kg reduced mean lung bacillary burden by an additional 0.69 log10 (P=0.01) relative to SPaO alone. As early as 2 weeks post-treatment initiation, SnMP adjunctive therapy differentially altered the expression of pro-inflammatory cytokine genes, and CD38, a marker of M1 macrophages. Next, we evaluated the sterilizing potential of SnMP adjunctive therapy in a mouse model of microbiological relapse. After 6 weeks of treatment, SPaO + SnMP 10 mg/kg reduced lung bacterial burdens to 0.71 ± 0.23 log10 CFU, a 0.78 log-fold greater decrease in lung CFU compared to SpaO alone (P=0.005). However, adjunctive SnMP did not reduce microbiological relapse rates after 5 or 6 weeks of treatment. SnMP was well tolerated and did not significantly alter gross or histological lung pathology. SnMP is a promising HDT candidate requiring further study in combination with regimens for drug-resistant TB.
Collapse
Affiliation(s)
- Jennie Ruelas Castillo
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pranita Neupane
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Styliani Karanika
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stefanie Krug
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Darla Quijada
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew Garcia
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel Ayeh
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Addis Yilma
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Diego L. Costa
- Departmento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Petros C. Karakousis
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
14
|
da Silva Graça Amoras E, de Morais TG, do Nascimento Ferreira R, Gomes STM, de Sousa FDM, de Paula Souza I, Ishak R, Vallinoto ACR, Queiroz MAF. Association of Cytokine Gene Polymorphisms and Their Impact on Active and Latent Tuberculosis in Brazil's Amazon Region. Biomolecules 2023; 13:1541. [PMID: 37892223 PMCID: PMC10605732 DOI: 10.3390/biom13101541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Some genetic variations in cytokine genes can alter their expression and influence the evolution of Mycobacterium tuberculosis (Mtb) infection. This study aimed to investigate the association of polymorphisms in cytokine genes and variability in plasma levels of cytokines with the development of tuberculosis (TB) and latent tuberculosis infection (LTBI). Blood samples from 245 patients with TB, 80 with LTBI, and healthy controls (n = 100) were included. Genotyping of the IFNG +874A/T, IL6 -174G/C, IL4 -590C/T, and IL10 -1082A/G polymorphisms was performed by real-time PCR, and cytokine levels were determined by flow cytometry. Higher frequencies of genotypes AA (IFNG +874A/T), GG (IL6 -174G/C), TT (IL4 -590C/T), and GG (IL10 -1082A/G) were associated with an increased risk of TB compared to that of LTBI (p = 0.0027; p = 0.0557; p = 0.0286; p = 0.0361, respectively) and the control (p = <0.0001, p = 0.0021; p = 0.01655; p = 0.0132, respectively). In combination, the A allele for IFNG +874A/T and the T allele for IL4 -590C/T were associated with a higher chance of TB (p = 0.0080; OR = 2.753 and p < 0.0001; OR = 3.273, respectively). The TB group had lower levels of IFN-γ and higher concentrations of IL-6, IL-4, and IL-10. Cytokine levels were different between the genotypes based on the polymorphisms investigated (p < 0.05). The genotype and wild-type allele for IFNG +874A/T and the genotype and polymorphic allele for IL4 -590C/T appear to be more relevant in the context of Mtb infection, which has been associated with the development of TB among individuals infected by the bacillus and with susceptibility to active infection but not with susceptibility to latent infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maria Alice Freitas Queiroz
- Virus Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.d.S.G.A.); (T.G.d.M.); (R.d.N.F.); (S.T.M.G.); (F.D.M.d.S.); (I.d.P.S.); (R.I.); (A.C.R.V.)
| |
Collapse
|
15
|
Gail DP, Suzart VG, Du W, Kaur Sandhu A, Jarvela J, Nantongo M, Mwebaza I, Panigrahi S, Freeman ML, Canaday DH, Boom WH, Silver RF, Carpenter SM. Mycobacterium tuberculosis impairs human memory CD4 + T cell recognition of M2 but not M1-like macrophages. iScience 2023; 26:107706. [PMID: 37694142 PMCID: PMC10485162 DOI: 10.1016/j.isci.2023.107706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/24/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023] Open
Abstract
Direct recognition of Mycobacterium tuberculosis (Mtb)-infected cells is required for protection by CD4+ T cells. While impaired T cell recognition of Mtb-infected macrophages was demonstrated in mice, data are lacking for humans. Using T cells and monocyte-derived macrophages (MDMs) from individuals with latent Mtb infection (LTBI), we quantified the frequency of memory CD4+ T cell activation in response to autologous MDMs infected with virulent Mtb. We observed robust T cell activation in response to Mtb infection of M1-like macrophages differentiated using GM-CSF, while M2-like macrophages differentiated using M-CSF were poorly recognized. However, non-infected GM-CSF and M-CSF MDMs loaded with exogenous antigens elicited similar CD4+ T cell activation. IL-10 was preferentially secreted by infected M-CSF MDMs, and neutralization improved T cell activation. These results suggest that preferential infection of macrophages with an M2-like phenotype limits T cell-mediated protection against Mtb. Vaccine development should focus on T cell recognition of Mtb-infected macrophages.
Collapse
Affiliation(s)
- Daniel P. Gail
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Vinicius G. Suzart
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Biomedical Sciences Training Program, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Weinan Du
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Avinaash Kaur Sandhu
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Biomedical Sciences Training Program, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jessica Jarvela
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Medicine, The Louis Stokes Cleveland V.A. Medical Center, Cleveland, OH 44106, USA
| | - Mary Nantongo
- Biomedical Sciences Training Program, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ivan Mwebaza
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Biomedical Sciences Training Program, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Soumya Panigrahi
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Biomedical Sciences Training Program, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - David H. Canaday
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Medicine, The Louis Stokes Cleveland V.A. Medical Center, Cleveland, OH 44106, USA
| | - W. Henry Boom
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Biomedical Sciences Training Program, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44139, USA
| | - Richard F. Silver
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Medicine, The Louis Stokes Cleveland V.A. Medical Center, Cleveland, OH 44106, USA
| | - Stephen M. Carpenter
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Biomedical Sciences Training Program, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44139, USA
| |
Collapse
|
16
|
Wang X, Wang X, Lei X, He Y, Xiao T. Photodynamic therapy: a new approach to the treatment of Nontuberculous Mycobacterial skin and soft tissue infections. Photodiagnosis Photodyn Ther 2023; 43:103645. [PMID: 37270047 DOI: 10.1016/j.pdpdt.2023.103645] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
Nontuberculous mycobacterial skin and soft tissue infections are rising and are causing social concern due to the growth of cosmetic dermatology and immune-compromised populations. For the treatment of nontuberculous mycobacteria, several novel strategies have been investigated. One of them, photodynamic therapy, is a recently developed therapeutic strategy that has shown promise in managing nontuberculous mycobacterial skin and soft tissue infections. In this review, we first present an overview of the current status of the therapy and then summarize and analyze the cases of photodynamic therapy used to treat nontuberculous mycobacterial skin and soft tissue infections. We also discussed the feasibility of photodynamic therapy for treating nontuberculous mycobacterial skin soft tissue infections and the related mechanisms, providing a potential new option for clinical treatment.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, CN
| | - Xiaoyu Wang
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, CN
| | - Xia Lei
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, CN.
| | - Yongqing He
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, CN
| | - Tianzhen Xiao
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, CN
| |
Collapse
|
17
|
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
|
18
|
Zhang QA, Ma S, Li P, Xie J. The dynamics of Mycobacterium tuberculosis phagosome and the fate of infection. Cell Signal 2023; 108:110715. [PMID: 37192679 DOI: 10.1016/j.cellsig.2023.110715] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/25/2023] [Accepted: 05/12/2023] [Indexed: 05/18/2023]
Abstract
Phagosomes are vesicles produced by phagocytosis of phagocytes, which are crucial in immunity against Mycobacterium tuberculosis (Mtb) infection. After the phagocyte ingests the pathogen, it activates the phagosomes to recruit a series of components and process proteins, to phagocytose, degrade and kill Mtb. Meanwhile, Mtb can resist acid and oxidative stress, block phagosome maturation, and manipulate host immune response. The interaction between Mtb and phagocytes leads to the outcome of infection. The dynamic of this process can affect the cell fate. This article mainly reviews the development and maturation of phagosomes, as well as the dynamics and modifications of Mtb effectors and phagosomes components, and new diagnostic and therapeutic markers involved in phagosomes.
Collapse
Affiliation(s)
- Qi-Ao Zhang
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Shaying Ma
- Chongqing Emergency Medical Center, Chongqing the Fourth Hospital, Jiankang Road, Yuzhong, Chongqing 400014, China
| | - Peibo Li
- Chongqing Public Health Medical Center, Chongqing, China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China; Chongqing Public Health Medical Center, Chongqing, China.
| |
Collapse
|
19
|
Dallmann-Sauer M, Xu YZ, da Costa ALF, Tao S, Gomes TA, Prata RBDS, Correa-Macedo W, Manry J, Alcaïs A, Abel L, Cobat A, Fava VM, Pinheiro RO, Lara FA, Probst CM, Mira MT, Schurr E. Allele-dependent interaction of LRRK2 and NOD2 in leprosy. PLoS Pathog 2023; 19:e1011260. [PMID: 36972292 PMCID: PMC10079233 DOI: 10.1371/journal.ppat.1011260] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/06/2023] [Accepted: 03/02/2023] [Indexed: 03/29/2023] Open
Abstract
Leprosy, caused by Mycobacterium leprae, rarely affects children younger than 5 years. Here, we studied a multiplex leprosy family that included monozygotic twins aged 22 months suffering from paucibacillary leprosy. Whole genome sequencing identified three amino acid mutations previously associated with Crohn’s disease and Parkinson’s disease as candidate variants for early onset leprosy: LRRK2 N551K, R1398H and NOD2 R702W. In genome-edited macrophages, we demonstrated that cells expressing the LRRK2 mutations displayed reduced apoptosis activity following mycobacterial challenge independently of NOD2. However, employing co-immunoprecipitation and confocal microscopy we showed that LRRK2 and NOD2 proteins interacted in RAW cells and monocyte-derived macrophages, and that this interaction was substantially reduced for the NOD2 R702W mutation. Moreover, we observed a joint effect of LRRK2 and NOD2 variants on Bacillus Calmette-Guérin (BCG)-induced respiratory burst, NF-κB activation and cytokine/chemokine secretion with a strong impact for the genotypes found in the twins consistent with a role of the identified mutations in the development of early onset leprosy.
Collapse
Affiliation(s)
- Monica Dallmann-Sauer
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
- Departments of Human Genetics and Medicine, Faculty of Medicine and Health Science, McGill University; Montreal, Canada
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná; Curitiba, Brazil
| | - Yong Zhong Xu
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
| | - Ana Lúcia França da Costa
- Department of Specialized Medicine, Health Sciences Center, Federal University of Piauí; Teresina, Brazil
| | - Shao Tao
- Division of Experimental Medicine, Faculty of Medicine, McGill University; Montreal, Canada
- The Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre; Montreal, Canada
| | - Tiago Araujo Gomes
- Laboratory of Cellular Microbiology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation; Rio de Janeiro, Brazil
| | | | - Wilian Correa-Macedo
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
- Department of Biochemistry, Faculty of Medicine and Health Science, McGill University; Montreal, Canada
| | - Jérémy Manry
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
| | - Alexandre Alcaïs
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, United States of America
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, United States of America
| | - Vinicius M. Fava
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
| | - Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation; Rio de Janeiro, Brazil
| | - Flavio Alves Lara
- Laboratory of Cellular Microbiology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation; Rio de Janeiro, Brazil
| | - Christian M. Probst
- Laboratory of Systems and Molecular Biology of Trypanosomatids, Instituto Carlos Chagas; FIOCRUZ, Curitiba, Brazil
| | - Marcelo T. Mira
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná; Curitiba, Brazil
- * E-mail: (M.T.M); (E.S.)
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
- Departments of Human Genetics and Medicine, Faculty of Medicine and Health Science, McGill University; Montreal, Canada
- Department of Biochemistry, Faculty of Medicine and Health Science, McGill University; Montreal, Canada
- * E-mail: (M.T.M); (E.S.)
| |
Collapse
|
20
|
Gollnick H, Barber J, Wilkinson RJ, Newton S, Garg A. IL-27 inhibits anti- Mycobacterium tuberculosis innate immune activity of primary human macrophages. Tuberculosis (Edinb) 2023; 139:102326. [PMID: 36863206 PMCID: PMC10052773 DOI: 10.1016/j.tube.2023.102326] [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: 06/22/2022] [Revised: 02/12/2023] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) is an intracellular pathogen that primarily infects macrophages. Despite a robust anti-mycobacterial response, many times macrophages are unable to control M. tuberculosis. The purpose of this study was to investigate the mechanism by which the immunoregulatory cytokine IL-27 inhibits the anti-mycobacterial activity of primary human macrophages. We found concerted production of IL-27 and anti-mycobacterial cytokines by M. tuberculosis-infected macrophages in a toll-like receptor (TLR) dependent manner. Notably, IL-27 suppressed the production of anti-mycobacterial cytokines TNFα, IL-6, IL-1β, and IL-15 by M. tuberculosis-infected macrophages. IL-27 limits the anti-mycobacterial activity of macrophages by reducing Cyp27B, cathelicidin (LL-37), LC3B lipidation, and increasing IL-10 production. Furthermore, neutralizing both IL-27 and IL-10 increased the expression of proteins involved in LC3-associated phagocytosis (LAP) pathway for bacterial clearance, namely vacuolar-ATPase, NOX2, and RUN-domain containing protein RUBCN. These results implicate IL-27 is a prominent cytokine that impedes M. tuberculosis clearance.
Collapse
Affiliation(s)
- Hailey Gollnick
- College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jamie Barber
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Robert J Wilkinson
- Department of Infectious Diseases, Imperial College London, W12 0NN, United Kingdom; The Francis Crick Institute London, NW1 1AT, United Kingdom
| | - Sandra Newton
- Section of Pediatric Infectious Disease, Department of Infectious Disease, Imperial College London, W2 1PG, United Kingdom
| | - Ankita Garg
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
21
|
Parsons JB, Westgeest AC, Conlon BP, Fowler VG. Persistent Methicillin-Resistant Staphylococcus aureus Bacteremia: Host, Pathogen, and Treatment. Antibiotics (Basel) 2023; 12:455. [PMID: 36978320 PMCID: PMC10044482 DOI: 10.3390/antibiotics12030455] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a devastating pathogen responsible for a variety of life-threatening infections. A distinctive characteristic of this pathogen is its ability to persist in the bloodstream for several days despite seemingly appropriate antibiotics. Persistent MRSA bacteremia is common and is associated with poor clinical outcomes. The etiology of persistent MRSA bacteremia is a result of the complex interplay between the host, the pathogen, and the antibiotic used to treat the infection. In this review, we explore the factors related to each component of the host-pathogen interaction and discuss the clinical relevance of each element. Next, we discuss the treatment options and diagnostic approaches for the management of persistent MRSA bacteremia.
Collapse
Affiliation(s)
- Joshua B. Parsons
- Department of Medicine, Division of Infectious Disease, Duke University Medical Center, Durham, NC 27710, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Annette C. Westgeest
- Department of Medicine, Division of Infectious Disease, Duke University Medical Center, Durham, NC 27710, USA
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Brian P. Conlon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Vance G. Fowler
- Department of Medicine, Division of Infectious Disease, Duke University Medical Center, Durham, NC 27710, USA
- Duke Clinical Research Institute, Durham, NC 27710, USA
| |
Collapse
|
22
|
Peruhype-Magalhães V, de Araújo FF, de Morais Papini TF, Wendling APB, Campi-Azevedo AC, Coelho-Dos-Reis JG, de Almeida IN, do Valle Antonnelli LR, Amaral LR, de Souza Gomes M, Brito-de-Sousa JP, Elói-Santos SM, Augusto VM, Pretti Dalcolmo MM, Carneiro CM, Teixeira-Carvalho A, Martins-Filho OA. Serum biomarkers in patients with unilateral or bilateral active pulmonary tuberculosis: Immunological networks and promising diagnostic applications. Cytokine 2023; 162:156076. [PMID: 36417816 DOI: 10.1016/j.cyto.2022.156076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 10/08/2022] [Accepted: 10/14/2022] [Indexed: 11/21/2022]
Abstract
The present observational study was designed to characterize the integrative profile of serum soluble mediators to describe the immunological networks associated with clinical findings and identify putative biomarkers for diagnosis and prognosis of active tuberculosis. The study population comprises 163 volunteers, including 84 patients with active pulmonary tuberculosis/(TB), and 79 controls/(C). Soluble mediators were measured by multiplexed assay. Data analysis demonstrated that the levels of CCL3, CCL5, CXCL10, IL-1β, IL-6, IFN-γ, IL-1Ra, IL-4, IL-10, PDGF, VEGF, G-CSF, IL-7 were increased in TB as compared to C. Patients with bilateral pulmonary involvement/(TB-BI) exhibited higher levels of CXCL8, IL-6 and TNF with distinct biomarker signatures (CCL11, CCL2, TNF and IL-10) as compared to patients with unilateral infiltrates/(TB-UNI). Analysis of biomarker networks based in correlation power graph demonstrated small number of strong connections in TB and TB-BI. The search for biomarkers with relevant implications to understand the pathogenetic mechanisms and useful as complementary diagnosis tool of active TB pointed out the excellent performance of single analysis of IL-6 or CXCL10 and the stepwise combination of IL-6 → CXCL10 (Accuracy = 84 %; 80 % and 88 %, respectively). Together, our finding demonstrated that immunological networks of serum soluble biomarkers in TB patients differ according to the unilateral or bilateral pulmonary involvement and may have relevant implications to understand the pathogenetic mechanisms involved in the clinical outcome of Mtb infection.
Collapse
Affiliation(s)
- Vanessa Peruhype-Magalhães
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Fortes de Araújo
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Tatiane Figueiredo de Morais Papini
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil; Pós-graduação em Ciências Farmacêuticas (CIPHARMA), Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Ana Paula Barbosa Wendling
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Carolina Campi-Azevedo
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Jordana Grazziela Coelho-Dos-Reis
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Isabela Neves de Almeida
- Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Lis Ribeiro do Valle Antonnelli
- Laboratório de Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou - FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| | - Laurence Rodrigues Amaral
- Rede Multidisciplinar de Pesquisa, Ciência e Tecnologia, Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Campus Patos de Minas, Patos de Minas, MG, Brazil
| | - Matheus de Souza Gomes
- Rede Multidisciplinar de Pesquisa, Ciência e Tecnologia, Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Campus Patos de Minas, Patos de Minas, MG, Brazil
| | - Joaquim Pedro Brito-de-Sousa
- Pós-graduação em Imunologia e Parasitologia Aplicadas (PPIPA), Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Silvana Maria Elói-Santos
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil; Departamento de Propedêutica Complementar, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Valéria Maria Augusto
- Departamento de Propedêutica Complementar, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Margareth Maria Pretti Dalcolmo
- Escola Nacional de Saúde Pública, Centro de Referência Professor Hélio Fraga, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cláudia Martins Carneiro
- Pós-graduação em Ciências Farmacêuticas (CIPHARMA), Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil; Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Andréa Teixeira-Carvalho
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil.
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
23
|
Selvapandiyan A, Puri N, Kumar P, Alam A, Ehtesham NZ, Griffin G, Hasnain SE. Zooming in on common immune evasion mechanisms of pathogens in phagolysosomes: potential broad-spectrum therapeutic targets against infectious diseases. FEMS Microbiol Rev 2023; 47:6780197. [PMID: 36309472 DOI: 10.1093/femsre/fuac041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 01/19/2023] Open
Abstract
The intracellular viral, bacterial, or parasitic pathogens evade the host immune challenges to propagate and cause fatal diseases. The microbes overpower host immunity at various levels including during entry into host cells, phagosome formation, phagosome maturation, phagosome-lysosome fusion forming phagolysosomes, acidification of phagolysosomes, and at times after escape into the cytosol. Phagolysosome is the final organelle in the phagocyte with sophisticated mechanisms to degrade the pathogens. The immune evasion strategies by the pathogens include the arrest of host cell apoptosis, decrease in reactive oxygen species, the elevation of Th2 anti-inflammatory response, avoidance of autophagy and antigen cross-presentation pathways, and escape from phagolysosomal killing. Since the phagolysosome organelle in relation to infection/cure is seldom discussed in the literature, we summarize here the common host as well as pathogen targets manipulated or utilized by the pathogens established in phagosomes and phagolysosomes, to hijack the host immune system for their benefit. These common molecules or pathways can be broad-spectrum therapeutic targets for drug development for intervention against infectious diseases caused by different intracellular pathogens.
Collapse
Affiliation(s)
| | - Niti Puri
- Cellular and Molecular Immunology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pankaj Kumar
- Department of Biochemistry, Jamia Hamdard, New Delhi, 110062, India.,Centre for Tuberculosis Research, Department of Medicine, Johns Hopkins University, Baltimore, MD, 21218, United States
| | - Anwar Alam
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India.,Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India
| | - Nasreen Zafar Ehtesham
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
| | - George Griffin
- Department of Cellular and Molecular Medicine, St. George's University of London, London, SW17 0RE, United Kingdom
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India.,Department of Life Science, School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, 201310, India
| |
Collapse
|
24
|
Feng Y, Zeng Q, Qiu Y, Li D, Shi D. Successful application of photodynamic therapy for skin infection caused by Corynespora cassiicola in an immunosuppressed patient and literature review. Photodiagnosis Photodyn Ther 2023; 41:103279. [PMID: 36632874 DOI: 10.1016/j.pdpdt.2023.103279] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023]
Abstract
Corynespora cassiicola (C. cassiicola) is a common plant pathogen but occasionally causes infections in human skin. Poor outcome and death in such cases were then often reported even after a long course of antifungal treatment. In the present case, the patient was a 54-year-old farmer who presented with a ten-month history of an ulcerative lesion in the right lower extremity after a minor skin trauma. The patient had been undergoing immunosuppressive treatment for nephrotic syndrome for 2 months before the onset of the skin lesion. The fungal isolate from the lesion was identified as C. cassiicola by morphology and molecular sequencing. The patient was treated with ALA-PDT 5 times at 10 days intervals. The lesion disappeared and has not recurred in the 8-month follow-up. To our knowledge, it is the first time ALA-PDT has been used for treatment of a skin lesion caused by C. cassiicola. A retrospective analysis of the literature found 9 cases of C. cassiicola infection. All 9 patients had a history of immunodeficient conditions and had experienced a long course of antifungal therapy. Despite the rigorous antifungal treatment, 2 patients with CARD9 gene deficiency ultimately died. Our case suggests that photodynamic therapy is a potentially beneficial approach for treatment of fungal infection in immunosuppressed patients who failed to respond to antifungal agents.
Collapse
Affiliation(s)
- Yahui Feng
- Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining 272067, Shandong Province, China
| | - Qian Zeng
- Department of Dermatology, Qufu People's Hospital, Jining 273199, Shandong, China
| | - Ying Qiu
- Department of Dermatology, Jining No.1 People's Hospital, Jining 272001, Shandong Province, China
| | - Dongmei Li
- Department of Microbiology/Immunology, Georgetown University Medical Center, Washington, DC 20057, United States
| | - Dongmei Shi
- Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining 272067, Shandong Province, China; Department of Dermatology, Jining No.1 People's Hospital, Jining 272001, Shandong Province, China.
| |
Collapse
|
25
|
Hasankhani A, Bahrami A, Mackie S, Maghsoodi S, Alawamleh HSK, Sheybani N, Safarpoor Dehkordi F, Rajabi F, Javanmard G, Khadem H, Barkema HW, De Donato M. In-depth systems biological evaluation of bovine alveolar macrophages suggests novel insights into molecular mechanisms underlying Mycobacterium bovis infection. Front Microbiol 2022; 13:1041314. [PMID: 36532492 PMCID: PMC9748370 DOI: 10.3389/fmicb.2022.1041314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/04/2022] [Indexed: 08/26/2023] Open
Abstract
Objective Bovine tuberculosis (bTB) is a chronic respiratory infectious disease of domestic livestock caused by intracellular Mycobacterium bovis infection, which causes ~$3 billion in annual losses to global agriculture. Providing novel tools for bTB managements requires a comprehensive understanding of the molecular regulatory mechanisms underlying the M. bovis infection. Nevertheless, a combination of different bioinformatics and systems biology methods was used in this study in order to clearly understand the molecular regulatory mechanisms of bTB, especially the immunomodulatory mechanisms of M. bovis infection. Methods RNA-seq data were retrieved and processed from 78 (39 non-infected control vs. 39 M. bovis-infected samples) bovine alveolar macrophages (bAMs). Next, weighted gene co-expression network analysis (WGCNA) was performed to identify the co-expression modules in non-infected control bAMs as reference set. The WGCNA module preservation approach was then used to identify non-preserved modules between non-infected controls and M. bovis-infected samples (test set). Additionally, functional enrichment analysis was used to investigate the biological behavior of the non-preserved modules and to identify bTB-specific non-preserved modules. Co-expressed hub genes were identified based on module membership (MM) criteria of WGCNA in the non-preserved modules and then integrated with protein-protein interaction (PPI) networks to identify co-expressed hub genes/transcription factors (TFs) with the highest maximal clique centrality (MCC) score (hub-central genes). Results As result, WGCNA analysis led to the identification of 21 modules in the non-infected control bAMs (reference set), among which the topological properties of 14 modules were altered in the M. bovis-infected bAMs (test set). Interestingly, 7 of the 14 non-preserved modules were directly related to the molecular mechanisms underlying the host immune response, immunosuppressive mechanisms of M. bovis, and bTB development. Moreover, among the co-expressed hub genes and TFs of the bTB-specific non-preserved modules, 260 genes/TFs had double centrality in both co-expression and PPI networks and played a crucial role in bAMs-M. bovis interactions. Some of these hub-central genes/TFs, including PSMC4, SRC, BCL2L1, VPS11, MDM2, IRF1, CDKN1A, NLRP3, TLR2, MMP9, ZAP70, LCK, TNF, CCL4, MMP1, CTLA4, ITK, IL6, IL1A, IL1B, CCL20, CD3E, NFKB1, EDN1, STAT1, TIMP1, PTGS2, TNFAIP3, BIRC3, MAPK8, VEGFA, VPS18, ICAM1, TBK1, CTSS, IL10, ACAA1, VPS33B, and HIF1A, had potential targets for inducing immunomodulatory mechanisms by M. bovis to evade the host defense response. Conclusion The present study provides an in-depth insight into the molecular regulatory mechanisms behind M. bovis infection through biological investigation of the candidate non-preserved modules directly related to bTB development. Furthermore, several hub-central genes/TFs were identified that were significant in determining the fate of M. bovis infection and could be promising targets for developing novel anti-bTB therapies and diagnosis strategies.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Shayan Mackie
- Faculty of Science, Earth Sciences Building, University of British Columbia, Vancouver, BC, Canada
| | - Sairan Maghsoodi
- Faculty of Paramedical Sciences, Kurdistan University of Medical Sciences, Kurdistan, Iran
| | - Heba Saed Kariem Alawamleh
- Department of Basic Scientific Sciences, AL-Balqa Applied University, AL-Huson University College, AL-Huson, Jordan
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Farhad Safarpoor Dehkordi
- Halal Research Center of IRI, FDA, Tehran, Iran
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Fatemeh Rajabi
- Department of Agronomy and Plant Breeding, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Hosein Khadem
- Department of Agronomy and Plant Breeding, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Marcos De Donato
- Regional Department of Bioengineering, Tecnológico de Monterrey, Monterrey, Mexico
| |
Collapse
|
26
|
Enhanced immunogenicity of Mycobacterium bovis BCG through CRISPRi mediated depletion of AftC. Cell Surf 2022; 8:100088. [PMID: 36405350 PMCID: PMC9651938 DOI: 10.1016/j.tcsw.2022.100088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis causes the disease tuberculosis and affects a third of the world’s population. The recent COVID-19 pandemic exacerbated the situation with a projected 27% increase in tuberculosis related deaths. M. tuberculosis has an elaborate cell wall consisting of peptidoglycan, arabinogalactan and mycolic acids which shield the bacilli from the toxic bactericidal milieu within phagocytes. Amongst, the numerous glycosyltransferase enzymes involved in mycobacterial cell wall biosynthesis, arabinofuranosyltransferase C (aftC) is responsible for the branching of the arabinan domain in both arabinogalactan and lipoarabinomannan. Using Clustered Regularly Interspaced Short Palindromic Repeats interference (CRISPRi) we have generated aftC knockdowns in Mycobacterium bovis BCG and demonstrated the generation of a truncated, immunogenic lipoarabinomannan within its cell envelope. The aftC depleted BCG mutants were unable to form characteristic mycobacterial pellicular biofilms and elicit a potent immunostimulatory phenotype compared to wild type M. bovis BCG in a THP1 cell line. This study paves the way to further explore novel BCG mutants as promising vaccine boosters in preventing pulmonary tuberculosis.
Collapse
|
27
|
Nisa A, Kipper FC, Panigrahy D, Tiwari S, Kupz A, Subbian S. Different modalities of host cell death and their impact on Mycobacterium tuberculosis infection. Am J Physiol Cell Physiol 2022; 323:C1444-C1474. [PMID: 36189975 PMCID: PMC9662802 DOI: 10.1152/ajpcell.00246.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 11/22/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is the pathogen that causes tuberculosis (TB), a leading infectious disease of humans worldwide. One of the main histopathological hallmarks of TB is the formation of granulomas comprised of elaborately organized aggregates of immune cells containing the pathogen. Dissemination of Mtb from infected cells in the granulomas due to host and mycobacterial factors induces multiple cell death modalities in infected cells. Based on molecular mechanism, morphological characteristics, and signal dependency, there are two main categories of cell death: programmed and nonprogrammed. Programmed cell death (PCD), such as apoptosis and autophagy, is associated with a protective response to Mtb by keeping the bacteria encased within dead macrophages that can be readily phagocytosed by arriving in uninfected or neighboring cells. In contrast, non-PCD necrotic cell death favors the pathogen, resulting in bacterial release into the extracellular environment. Multiple types of cell death in the PCD category, including pyroptosis, necroptosis, ferroptosis, ETosis, parthanatos, and PANoptosis, may be involved in Mtb infection. Since PCD pathways are essential for host immunity to Mtb, therapeutic compounds targeting cell death signaling pathways have been experimentally tested for TB treatment. This review summarizes different modalities of Mtb-mediated host cell deaths, the molecular mechanisms underpinning host cell death during Mtb infection, and its potential implications for host immunity. In addition, targeting host cell death pathways as potential therapeutic and preventive approaches against Mtb infection is also discussed.
Collapse
Affiliation(s)
- Annuurun Nisa
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Franciele C Kipper
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Sangeeta Tiwari
- Department of Biological Sciences, Border Biomedical Research Center (BBRC), University of Texas, El Paso, Texas
| | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Townsville, Queensland, Australia
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
28
|
Simper JD, Perez E, Schlesinger LS, Azad AK. Resistance and Susceptibility Immune Factors at Play during Mycobacterium tuberculosis Infection of Macrophages. Pathogens 2022; 11:pathogens11101153. [PMID: 36297211 PMCID: PMC9611686 DOI: 10.3390/pathogens11101153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/28/2022] Open
Abstract
Tuberculosis (TB), caused by infection with Mycobacterium tuberculosis (M.tb), is responsible for >1.5 million deaths worldwide annually. Innate immune cells, especially macrophages, are the first to encounter M.tb, and their response dictates the course of infection. During infection, macrophages exert a variety of immune factors involved in either controlling or promoting the growth of M.tb. Research on this topic has been performed in both in vitro and in vivo animal models with discrepant results in some cases based on the model of study. Herein, we review macrophage resistance and susceptibility immune factors, focusing primarily on recent advances in the field. We include macrophage cellular pathways, bioeffector proteins and molecules, cytokines and chemokines, associated microbiological factors and bacterial strains, and host genetic factors in innate immune genes. Recent advances in mechanisms underlying macrophage resistance and susceptibility factors will aid in the successful development of host-directed therapeutics, a topic emphasized throughout this review.
Collapse
Affiliation(s)
- Jan D. Simper
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Department of Microbiology, Immunology and Molecular Genetics, UT Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Esteban Perez
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Translational Sciences Program, UT Health San Antonio Graduate School of Biomedical Sciences, San Antonio, TX 78229, USA
| | - Larry S. Schlesinger
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Correspondence: (L.S.S.); (A.K.A.); Tel.: +1-210-258-9578 (L.S.S.); +1-210-258-9467 (A.K.A.)
| | - Abul K. Azad
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Correspondence: (L.S.S.); (A.K.A.); Tel.: +1-210-258-9578 (L.S.S.); +1-210-258-9467 (A.K.A.)
| |
Collapse
|
29
|
Li W, Deng W, Zhang N, Peng H, Xu Y. Mycobacterium tuberculosis Rv2387 Facilitates Mycobacterial Survival by Silencing TLR2/p38/JNK Signaling. Pathogens 2022; 11:pathogens11090981. [PMID: 36145413 PMCID: PMC9504853 DOI: 10.3390/pathogens11090981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) can evade antimicrobial immunity and persist within macrophages by interfering with multiple host cellular functions through its virulence factors, causing latent tuberculosis. The Rv2387 protein has been identified as a putative effector that potentially participates in Mtb pathogenicity. To explore the role of the Rv2387 protein in host–mycobacteria interactions, we established recombinant M. smegmatis strains and RAW264.7 cell lines that stably express the Rv2387 protein. We found that this protein suppresses mycobacteria infection-induced macrophage apoptosis by inactivating caspase-3/-8, thus facilitating the intracellular survival of mycobacteria. In addition, Rv2387 inhibits the production of inflammatory cytokines in macrophages by specifically suppressing TLR2-dependent stimulation of p38 and JNK MAPK pathways. Moreover, we further determined that the Rv2387 protein conferred a growth advantage over recombinant M. smegmatis and suppressed the inflammatory response in a mouse infection model. Overall, these data suggested that Rv2387 facilitates mycobacteria to escape host immunity and might be an essential virulence factor in Mtb.
Collapse
Affiliation(s)
- Wu Li
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
- Correspondence: (W.L.); (Y.X.)
| | - Wanyan Deng
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nan Zhang
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
| | - Huijuan Peng
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
| | - Yi Xu
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- Correspondence: (W.L.); (Y.X.)
| |
Collapse
|
30
|
Bahlool AZ, Fattah S, O’Sullivan A, Cavanagh B, MacLoughlin R, Keane J, O’Sullivan MP, Cryan SA. Development of Inhalable ATRA-Loaded PLGA Nanoparticles as Host-Directed Immunotherapy against Tuberculosis. Pharmaceutics 2022; 14:pharmaceutics14081745. [PMID: 36015371 PMCID: PMC9415714 DOI: 10.3390/pharmaceutics14081745] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/11/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Developing new effective treatment strategies to overcome the rise in multi-drug resistant tuberculosis cases (MDR-TB) represents a global challenge. A host-directed therapy (HDT), acting on the host immune response rather than Mtb directly, could address these resistance issues. We developed an HDT for targeted TB treatment, using All Trans Retinoic Acid (ATRA)-loaded nanoparticles (NPs) that are suitable for nebulization. Efficacy studies conducted on THP-1 differentiated cells infected with the H37Ra avirulent Mycobacterium tuberculosis (Mtb) strain, have shown a dose-dependent reduction in H37Ra growth as determined by the BACT/ALERT® system. Confocal microscopy images showed efficient and extensive cellular delivery of ATRA-PLGA NPs into THP-1-derived macrophages. A commercially available vibrating mesh nebulizer was used to generate nanoparticle-loaded droplets with a mass median aerodynamic diameter of 2.13 μm as measured by cascade impaction, and a volumetric median diameter of 4.09 μm as measured by laser diffraction. In an adult breathing simulation experiment, 65.1% of the ATRA PLGA-NP dose was inhaled. This targeted inhaled HDT could offer a new adjunctive TB treatment option that could enhance current dosage regimens leading to better patient prognosis and a decreasing incidence of MDR-TB.
Collapse
Affiliation(s)
- Ahmad Z. Bahlool
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, D02 YN77 Dublin, Ireland
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, D02 YN77 Dublin, Ireland
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James’s Hospital, Trinity College Dublin, The University of Dublin, D08 9WRT Dublin, Ireland
| | - Sarinj Fattah
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, D02 YN77 Dublin, Ireland
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, D02 YN77 Dublin, Ireland
| | - Andrew O’Sullivan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, D02 YN77 Dublin, Ireland
- Research and Development, Science and Emerging Technologies, Aerogen Ltd., Galway Business Park, Dangan, H91 HE94 Galway, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland RCSI, D02 YN77 Dublin, Ireland
| | - Ronan MacLoughlin
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, D02 YN77 Dublin, Ireland
- Research and Development, Science and Emerging Technologies, Aerogen Ltd., Galway Business Park, Dangan, H91 HE94 Galway, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James’s Hospital, Trinity College Dublin, The University of Dublin, D08 9WRT Dublin, Ireland
| | - Mary P. O’Sullivan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James’s Hospital, Trinity College Dublin, The University of Dublin, D08 9WRT Dublin, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, D02 YN77 Dublin, Ireland
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, D02 YN77 Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI and Trinity College Dublin, D02 PN40 Dublin, Ireland
- SFI Centre for Research in Medical Devices (CÚRAM), NUIG & RCSI, H91 W2TY Galway, Ireland
- Correspondence:
| |
Collapse
|
31
|
Herrera MT, Guzmán-Beltrán S, Bobadilla K, Santos-Mendoza T, Flores-Valdez MA, Gutiérrez-González LH, González Y. Human Pulmonary Tuberculosis: Understanding the Immune Response in the Bronchoalveolar System. Biomolecules 2022; 12:biom12081148. [PMID: 36009042 PMCID: PMC9405639 DOI: 10.3390/biom12081148] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Mycobacterium tuberculosis, the causal agent of one of the most devastating infectious diseases worldwide, can evade or modulate the host immune response and remain dormant for many years. In this review, we focus on identifying the local immune response induced in vivo by M. tuberculosis in the lungs of patients with active tuberculosis by analyzing data from untouched cells from bronchoalveolar lavage fluid (BALF) or exhaled breath condensate (EBC) samples. The most abundant resident cells in patients with active tuberculosis are macrophages and lymphocytes, which facilitate the recruitment of neutrophils. The cellular response is characterized by an inflammatory state and oxidative stress produced mainly by macrophages and T lymphocytes. In the alveolar microenvironment, the levels of cytokines such as interleukins (IL), chemokines, and matrix metalloproteinases (MMP) are increased compared with healthy patients. The production of cytokines such as interferon (IFN)-γ and IL-17 and specific immunoglobulin (Ig) A and G against M. tuberculosis indicate that the adaptive immune response is induced despite the presence of a chronic infection. The role of epithelial cells, the processing and presentation of antigens by macrophages and dendritic cells, as well as the role of tissue-resident memory T cells (Trm) for in situ vaccination remains to be understood.
Collapse
Affiliation(s)
- María Teresa Herrera
- Department of Microbiology, National Institute for Respiratory Diseases Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Silvia Guzmán-Beltrán
- Department of Microbiology, National Institute for Respiratory Diseases Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Karen Bobadilla
- Laboratory of Transcriptomics and Molecular Immunology, National Institute for Respiratory Diseases Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Teresa Santos-Mendoza
- Laboratory of Transcriptomics and Molecular Immunology, National Institute for Respiratory Diseases Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Mario Alberto Flores-Valdez
- Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C., Guadalajara 44270, Mexico
| | - Luis Horacio Gutiérrez-González
- Laboratory of Transcriptomics and Molecular Immunology, National Institute for Respiratory Diseases Ismael Cosío Villegas, Mexico City 14080, Mexico
- Correspondence: (L.H.G.-G.); (Y.G.); Tel.: +52-55-5487-1700 (ext. 5117) (Y.G.)
| | - Yolanda González
- Department of Microbiology, National Institute for Respiratory Diseases Ismael Cosío Villegas, Mexico City 14080, Mexico
- Correspondence: (L.H.G.-G.); (Y.G.); Tel.: +52-55-5487-1700 (ext. 5117) (Y.G.)
| |
Collapse
|
32
|
Gupta M, Srikrishna G, Klein SL, Bishai WR. Genetic and hormonal mechanisms underlying sex-specific immune responses in tuberculosis. Trends Immunol 2022; 43:640-656. [PMID: 35842266 PMCID: PMC9344469 DOI: 10.1016/j.it.2022.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/06/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022]
Abstract
Tuberculosis (TB), the world's deadliest bacterial infection, afflicts more human males than females, with a male/female (M/F) ratio of 1.7. Sex disparities in TB prevalence, pathophysiology, and clinical manifestations are widely reported, but the underlying biological mechanisms remain largely undefined. This review assesses epidemiological data on sex disparity in TB, as well as possible underlying hormonal and genetic mechanisms that might differentially modulate innate and adaptive immune responses in males and females, leading to sex differences in disease susceptibility. We consider whether this sex disparity can be extended to the efficacy of vaccines and discuss novel animal models which may offer mechanistic insights. A better understanding of the biological factors underpinning sex-related immune responses in TB may enable sex-specific personalized therapies for TB.
Collapse
|
33
|
Arish M, Naz F. Sphingosine-1-phosphate receptors 2 and 3 reprogram resting human macrophages into M1 phenotype following mycobacteria infection. CURRENT RESEARCH IN IMMUNOLOGY 2022; 3:110-117. [PMID: 35676924 PMCID: PMC9168381 DOI: 10.1016/j.crimmu.2022.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/14/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022] Open
Abstract
Mycobacteria tuberculosis (M.tb) the causative agent for tuberculosis has been accredited for a high rate of morbidity and mortality worldwide. The rise in MDR and XDR cases has further created new obstacles in achieving the "End TB Strategy", which is aimed for 2035. In this article, we have demonstrated the potential of sphingosine-1-phosphate (S1P) analogs in providing an anti-mycobacterial effector response by altering macrophage polarity into M1. Among S1PR1 and S1PR3 analogs, S1PR2 analogs proficiently favor selective polarization of infected human macrophages into M1 phenotypes, marked by increased expression of M1 markers and decreased M2 markers. Furthermore, S1PR1-3 analogs treated macrophages were also able to decrease the secretion of anti-inflammatory cytokine IL-10 and can induce NO secretion in infected macrophages. Lastly, only S1PR2-3 analogs were able to restrict the growth of mycobacteria in human macrophages. Taken together our study reflects the potential of S1PR2-3 analogs in providing host defenses following mycobacterial infection by favoring M1 macrophage polarization.
Collapse
Affiliation(s)
- Mohd Arish
- JH-Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Farha Naz
- Centre of Interdisciplinary Research in Basic Science (CIRBSc), Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
34
|
Bahlool AZ, Grant C, Cryan SA, Keane J, O'Sullivan MP. All trans retinoic acid as a host-directed immunotherapy for tuberculosis. CURRENT RESEARCH IN IMMUNOLOGY 2022; 3:54-72. [PMID: 35496824 PMCID: PMC9040133 DOI: 10.1016/j.crimmu.2022.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/11/2022] [Accepted: 03/22/2022] [Indexed: 12/22/2022] Open
Abstract
Tuberculosis (TB) is the top bacterial infectious disease killer and one of the top ten causes of death worldwide. The emergence of strains of multiple drug-resistant tuberculosis (MDR-TB) has pushed our available stock of anti-TB agents to the limit of effectiveness. This has increased the urgent need to develop novel treatment strategies using currently available resources. An adjunctive, host-directed therapy (HDT) designed to act on the host, instead of the bacteria, by boosting the host immune response through activation of intracellular pathways could be the answer. The integration of multidisciplinary approaches of repurposing currently FDA-approved drugs, with a targeted drug-delivery platform is a very promising option to reduce the long timeline associated with the approval of new drugs - time that cannot be afforded given the current levels of morbidity and mortality associated with TB infection. The deficiency of vitamin A has been reported to be highly associated with the increased susceptibility of TB. All trans retinoic acid (ATRA), the active metabolite of vitamin A, has proven to be very efficacious against TB both in vitro and in vivo. In this review, we discuss and summarise the importance of vitamin A metabolites in the fight against TB and what is known regarding the molecular mechanisms of ATRA as a host-directed therapy for TB including its effect on macrophages cytokine profile and cellular pathways. Furthermore, we focus on the issues behind why previous clinical trials with vitamin A supplementation have failed, and how these issues might be overcome.
Collapse
Affiliation(s)
- Ahmad Z. Bahlool
- School of Pharmacy and Biomolecular Sciences (PBS), Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, Ireland
- Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, Ireland
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Conor Grant
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences (PBS), Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, Ireland
- Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
- SFI Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Mary P. O'Sullivan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| |
Collapse
|
35
|
Gupta R, Pandey M, Pandey AK, Tiwari PK, Amrathlal RS. Novel genetic polymorphisms identified in the clinical isolates of Mycobacterium tuberculosis PE_PGRS33 gene modulate cytokines expression and promotes survival in macrophages. J Infect Public Health 2022; 15:245-254. [DOI: 10.1016/j.jiph.2022.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/15/2021] [Accepted: 01/07/2022] [Indexed: 01/15/2023] Open
|
36
|
Bai KJ, Tung NT, Hsiao TC, Chen TL, Chung KF, Ho SC, Tsai CY, Chen JK, Lee CN, Lee KY, Chang CC, Chen TT, Feng PH, Chen KY, Su CL, Thao HNX, Dung HB, Thuy TPC, Lee YL, Chuang HC. Associations between lung-deposited dose of particulate matter and culture-positive pulmonary tuberculosis pleurisy. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:6140-6150. [PMID: 34448140 DOI: 10.1007/s11356-021-16008-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/13/2021] [Indexed: 06/13/2023]
Abstract
Epidemiological studies identified the relationship between air pollution and pulmonary tuberculosis. Effects of lung-deposited dose of particulate matter (PM) on culture-positive pulmonary tuberculosis remain unclear. This study investigates the association between lung-deposited dose of PM and pulmonary tuberculosis pleurisy. A case-control study of subjects undergoing pleural effusion drainage of pulmonary tuberculosis (case) and chronic heart failure (control) was conducted. Metals and biomarkers were quantified in the pleural effusion. The air pollution exposure was measured and PM deposition in the head, tracheobronchial, alveolar region, and total lung region was estimated by Multiple-path Particle Dosimetry (MPPD) Model. We performed multiple logistic regression to examine the associations of these factors with the risk of tuberculosis. We observed that 1-μg/m3 increase in PM10 was associated with 1.226-fold increased crude odds ratio (OR) of tuberculosis (95% confidence interval (CI): 1.023-1.469, p<0.05), 1-μg/m3 increase in PM2.5-10 was associated with 1.482-fold increased crude OR of tuberculosis (95% CI: 1.048-2.097, p < 0.05), 1-ppb increase in NO2 was associated with 1.218-fold increased crude OR of tuberculosis (95% CI: 1.025-1.447, p < 0.05), and 1-ppb increase in O3 was associated with 0.735-fold decreased crude OR of tuberculosis (95% CI: 0.542 0.995). We observed 1-μg/m3 increase in PM deposition in head and nasal region was associated with 1.699-fold increased crude OR of tuberculosis (95% CI: 1.065-2.711, p < 0.05), 1-μg/m3 increase in PM deposition in tracheobronchial region was associated with 1.592-fold increased crude OR of tuberculosis (95% CI: 1.095-2.313, p < 0.05), 1-μg/m3 increase in PM deposition in alveolar region was associated with 3.981-fold increased crude OR of tuberculosis (95% CI: 1.280-12.386, p < 0.05), and 1-μg/m3 increase in PM deposition in total lung was associated with 1.511-fold increased crude OR of tuberculosis (95% CI: 1.050-2.173, p < 0.05). The results indicate that particle deposition in alveolar region could cause higher risk of pulmonary tuberculosis pleurisy than deposition in other lung regions.
Collapse
Affiliation(s)
- Kuan-Jen Bai
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Nguyen Thanh Tung
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Otorhinolaryngology Department, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Tsai-Ling Chen
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Kian Fan Chung
- Faculty of Medicine, National Heart & Lung Institute, Imperial College London, London, UK
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Yu Tsai
- Department of Civil and Environmental Engineering, Imperial College London, London, UK
| | - Jen-Kun Chen
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Life Sciences and School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Nin Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Cheng Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chien-Ling Su
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | | | - Hoang Ba Dung
- Otorhinolaryngology Department, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Tran Phan Chung Thuy
- Otorhinolaryngology Department, Faculty of Medicine, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
37
|
Lundahl MLE, Mitermite M, Ryan DG, Case S, Williams NC, Yang M, Lynch RI, Lagan E, Lebre FM, Gorman AL, Stojkovic B, Bracken AP, Frezza C, Sheedy FJ, Scanlan EM, O'Neill LAJ, Gordon SV, Lavelle EC. Macrophage innate training induced by IL-4 and IL-13 activation enhances OXPHOS driven anti-mycobacterial responses. eLife 2022; 11:74690. [PMID: 36173104 PMCID: PMC9555863 DOI: 10.7554/elife.74690] [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: 10/13/2021] [Accepted: 09/28/2022] [Indexed: 01/05/2023] Open
Abstract
Macrophages are a highly adaptive population of innate immune cells. Polarization with IFNγ and LPS into the 'classically activated' M1 macrophage enhances pro-inflammatory and microbicidal responses, important for eradicating bacteria such as Mycobacterium tuberculosis. By contrast, 'alternatively activated' M2 macrophages, polarized with IL-4, oppose bactericidal mechanisms and allow mycobacterial growth. These activation states are accompanied by distinct metabolic profiles, where M1 macrophages favor near exclusive use of glycolysis, whereas M2 macrophages up-regulate oxidative phosphorylation (OXPHOS). Here, we demonstrate that activation with IL-4 and IL-13 counterintuitively induces protective innate memory against mycobacterial challenge. In human and murine models, prior activation with IL-4/13 enhances pro-inflammatory cytokine secretion in response to a secondary stimulation with mycobacterial ligands. In our murine model, enhanced killing capacity is also demonstrated. Despite this switch in phenotype, IL-4/13 trained murine macrophages do not demonstrate M1-typical metabolism, instead retaining heightened use of OXPHOS. Moreover, inhibition of OXPHOS with oligomycin, 2-deoxy glucose or BPTES all impeded heightened pro-inflammatory cytokine responses from IL-4/13 trained macrophages. Lastly, this work identifies that IL-10 attenuates protective IL-4/13 training, impeding pro-inflammatory and bactericidal mechanisms. In summary, this work provides new and unexpected insight into alternative macrophage activation states in the context of mycobacterial infection.
Collapse
Affiliation(s)
- Mimmi LE Lundahl
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland,School of Chemistry, Scanlan Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Morgane Mitermite
- School of Veterinary Medicine, UCD Veterinary Sciences Centre, University College DublinDublinIreland
| | - Dylan Gerard Ryan
- School of Biochemistry and Immunology, Inflammation Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland,Hutchison/MRC Research centre, MRC Cancer Unit, University of CambridgeCambridgeUnited Kingdom
| | - Sarah Case
- School of Biochemistry and Immunology, Macrophage Homeostasis Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Niamh C Williams
- School of Biochemistry and Immunology, Inflammation Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Ming Yang
- Hutchison/MRC Research centre, MRC Cancer Unit, University of CambridgeCambridgeUnited Kingdom
| | - Roisin I Lynch
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Eimear Lagan
- School of Genetics and Microbiology, Department of Genetics, Trinity College DublinDublinIreland
| | - Filipa M Lebre
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Aoife L Gorman
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Bojan Stojkovic
- School of Veterinary Medicine, UCD Veterinary Sciences Centre, University College DublinDublinIreland
| | - Adrian P Bracken
- School of Genetics and Microbiology, Department of Genetics, Trinity College DublinDublinIreland
| | - Christian Frezza
- Hutchison/MRC Research centre, MRC Cancer Unit, University of CambridgeCambridgeUnited Kingdom
| | - Frederick J Sheedy
- School of Biochemistry and Immunology, Macrophage Homeostasis Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Eoin M Scanlan
- School of Chemistry, Scanlan Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Luke AJ O'Neill
- School of Biochemistry and Immunology, Inflammation Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Stephen V Gordon
- School of Veterinary Medicine, UCD Veterinary Sciences Centre, University College DublinDublinIreland
| | - Ed C Lavelle
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| |
Collapse
|
38
|
Sontyana B, Shrivastava R, Battu S, Ghosh S, Mukhopadhyay S. Phagosome maturation and modulation of macrophage effector function by intracellular pathogens: target for therapeutics. Future Microbiol 2021; 17:59-76. [PMID: 34877879 DOI: 10.2217/fmb-2021-0101] [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: 11/21/2022] Open
Abstract
Macrophages are important cells that regulate various innate functions. Macrophages after engulfment of pathogens proceed for phagosome maturation and finally fuse with lysosomes to kill pathogens. Although pathogen degradation is one of the important functions of phagosomes, various immune-effector functions of macrophages are also dependent on the phagosome maturation process. This review discusses signaling processes regulating phagosome maturation as well as various effector functions of macrophages such as apoptosis, antigen presentation, autophagy and inflammasome that are dependent on the phagosome maturation process. It also discusses strategies adopted by various intracellular pathogens to counteract these functions to evade intracellular destruction mechanisms. These studies may give direction for the development of new therapeutics to control various intracellular infections.
Collapse
Affiliation(s)
- Brahmaji Sontyana
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rohini Shrivastava
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Srikanth Battu
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India
| | - Sudip Ghosh
- Molecular Biology Unit, ICMR-National Institute of Nutrition, Jamai Osmania PO, Hyderabad, 500007, Telangana, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India
| |
Collapse
|
39
|
Valdez HA, Marin Franco JL, Gorgojo JP, Alvarez Hayes J, Balboa L, Fernandez Lahore M, Sasiain MC, Rodriguez ME. Human macrophage polarization shapes B. pertussis intracellular persistence. J Leukoc Biol 2021; 112:173-184. [PMID: 34811790 DOI: 10.1002/jlb.4a0521-254r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/17/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
We previously demonstrated that Bordetella pertussis, the etiologic agent of whooping cough, is able to survive inside human macrophages. The aim of this study was to examine the influence of macrophage polarization in the development of B. pertussis intracellular infections. To this end, primary human monocytes were differentiated into M1, M2a, or M2c macrophages and further infected with B. pertussis. Infected M1 macrophages showed a proinflammatory response evidenced by the production of TNF-α, IL-12p70, and IL-6. Conversely, infection of M2a and M2c macrophages did not induce TNF-α, IL-12p70, nor IL-6 at any time postinfection but showed a significant increase of M2 markers, such as CD206, CD163, and CD209. Interestingly, anti-inflammatory cytokines, like IL-10 and TGF-β, were induced after infection in the 3 macrophage phenotypes. B. pertussis phagocytosis by M1 macrophages was lower than by M2 phenotypes, which may be ascribed to differences in the expression level of B. pertussis docking molecules on the surface of the different phenotypes. Intracellular bactericidal activity was found to be significantly higher in M1 than in M2a or M2c cells, but live bacteria were still detected within the 3 phenotypes at the late time points after infection. In summary, this study shows that intracellular B. pertussis is able to survive regardless of the macrophage activation program, but its intracellular survival proved higher in M2 compared with the M1 macrophages, being M2c the best candidate to develop into a niche of persistence for B. pertussis.
Collapse
Affiliation(s)
- Hugo A Valdez
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany.,CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Jose L Marin Franco
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Ciudad autónoma de Buenos Aires, Argentina
| | - Juan P Gorgojo
- CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Jimena Alvarez Hayes
- CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Luciana Balboa
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Ciudad autónoma de Buenos Aires, Argentina
| | | | - María C Sasiain
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Ciudad autónoma de Buenos Aires, Argentina
| | - Maria Eugenia Rodriguez
- CINDEFI (UNLP CONICET La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
40
|
Afriyie-Asante A, Dabla A, Dagenais A, Berton S, Smyth R, Sun J. Mycobacterium tuberculosis Exploits Focal Adhesion Kinase to Induce Necrotic Cell Death and Inhibit Reactive Oxygen Species Production. Front Immunol 2021; 12:742370. [PMID: 34745115 PMCID: PMC8564185 DOI: 10.3389/fimmu.2021.742370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/04/2021] [Indexed: 01/25/2023] Open
Abstract
Tuberculosis is a deadly, contagious respiratory disease that is caused by the pathogenic bacterium Mycobacterium tuberculosis (Mtb). Mtb is adept at manipulating and evading host immunity by hijacking alveolar macrophages, the first line of defense against inhaled pathogens, by regulating the mode and timing of host cell death. It is established that Mtb infection actively blocks apoptosis and instead induces necrotic-like modes of cell death to promote disease progression. This survival strategy shields the bacteria from destruction by the immune system and antibiotics while allowing for the spread of bacteria at opportunistic times. As such, it is critical to understand how Mtb interacts with host macrophages to manipulate the mode of cell death. Herein, we demonstrate that Mtb infection triggers a time-dependent reduction in the expression of focal adhesion kinase (FAK) in human macrophages. Using pharmacological perturbations, we show that inhibition of FAK (FAKi) triggers an increase in a necrotic form of cell death during Mtb infection. In contrast, genetic overexpression of FAK (FAK+) completely blocked macrophage cell death during Mtb infection. Using specific inhibitors of necrotic cell death, we show that FAK-mediated cell death during Mtb infection occurs in a RIPK1-depedent, and to a lesser extent, RIPK3-MLKL-dependent mechanism. Consistent with these findings, FAKi results in uncontrolled replication of Mtb, whereas FAK+ reduces the intracellular survival of Mtb in macrophages. In addition, we demonstrate that enhanced control of intracellular Mtb replication by FAK+ macrophages is a result of increased production of antibacterial reactive oxygen species (ROS) as inhibitors of ROS production restored Mtb burden in FAK+ macrophages to same levels as in wild-type cells. Collectively, our data establishes FAK as an important host protective response during Mtb infection to block necrotic cell death and induce ROS production, which are required to restrict the survival of Mtb.
Collapse
Affiliation(s)
- Afrakoma Afriyie-Asante
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Ankita Dabla
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Amy Dagenais
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Stefania Berton
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Robin Smyth
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Jim Sun
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
41
|
Kalra R, Tiwari D, Dkhar HK, Bhagyaraj E, Kumar R, Bhardwaj A, Gupta P. Host factors subverted by Mycobacterium tuberculosis: Potential targets for host directed therapy. Int Rev Immunol 2021; 42:43-70. [PMID: 34678117 DOI: 10.1080/08830185.2021.1990277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Despite new approaches in the diagnosis and treatment of tuberculosis (TB), it continues to be a major health burden. Several immunotherapies that potentiate the immune response have come up as adjuncts to drug therapies against drug resistant TB strains; however, there needs to be an urgent appraisal of host specific drug targets for improving their clinical management and to curtail disease progression. Presently, various host directed therapies (HDTs) exist (repurposed drugs, nutraceuticals, monoclonal antibodies and immunomodulatory agents), but these mostly address molecules that combat disease progression. AREAS COVERED The current review discusses major Mycobacterium tuberculosis (M. tuberculosis) survival paradigms inside the host and presents a plethora of host targets subverted by M. tuberculosis which can be further explored for future HDTs. The host factors unique to M. tuberculosis infection (in humans) have also been identified through an in-silico interaction mapping. EXPERT OPINION HDTs could become the next-generation adjunct therapies in order to counter antimicrobial resistance and virulence, as well as to reduce the duration of existing TB treatments. However, current scientific efforts are largely directed toward combatants rather than host molecules co-opted by M. tuberculosis for its survival. This might drive the immune system to a hyper-inflammatory condition; therefore, we emphasize that host factors subverted by M. tuberculosis, and their subsequent neutralization, must be considered for development of better HDTs.
Collapse
Affiliation(s)
- Rashi Kalra
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Drishti Tiwari
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Hedwin Kitdorlang Dkhar
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Ella Bhagyaraj
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Rakesh Kumar
- Bioinformatics Center, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Anshu Bhardwaj
- Bioinformatics Center, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Pawan Gupta
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
42
|
Fursov MV, Shitikov EA, Lagutkin DA, Fursova AD, Ganina EA, Kombarova TI, Grishenko NS, Rudnitskaya TI, Bespiatykh DA, Kolupaeva NV, Firstova VV, Domotenko LV, Panova AE, Vinokurov AS, Gushchin VA, Tkachuk AP, Vasilyeva IA, Potapov VD, Dyatlov IA. MDR and Pre-XDR Clinical Mycobacterium tuberculosis Beijing Strains: Assessment of Virulence and Host Cytokine Response in Mice Infectious Model. Microorganisms 2021; 9:1792. [PMID: 34442871 PMCID: PMC8400193 DOI: 10.3390/microorganisms9081792] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/05/2021] [Accepted: 08/18/2021] [Indexed: 11/16/2022] Open
Abstract
Mycobacterium tuberculosis Beijing genotype associated with drug resistance is a growing public health problem worldwide. The aim of this study was the assessment of virulence for C57BL/6 mice after infection by clinical M. tuberculosis strains 267/47 and 120/26, which belong to the modern sublineages B0/W148 and Central Asia outbreak of the Beijing genotype, respectively. The sublineages were identified by the analysis of the strains' whole-genomes. The strains 267/47 and 120/26 were characterized as agents of pre-extensively drug-resistant (pre-XDR) and multidrug-resistant (MDR) tuberculosis, respectively. Both clinical strains were slow-growing in 7H9 broth compared to the M. tuberculosis H37Rv strain. The survival rates of C57BL/6 mice infected by 267/47, 120/26, and H37Rv on the 150th day postinfection were 10%, 40%, and 70%, respectively. Mycobacterial load in the lungs, spleen, and liver was higher and histopathological changes were more expressed for mice infected by the 267/47 strain compared to those infected by the 120/26 and H37Rv strains. The cytokine response in the lungs of C57BL/6 mice after infection with the 267/47, 120/26, and H37Rv strains was different. Notably, proinflammatory cytokine genes Il-1α, Il-6, Il-7, and Il-17, as well as anti-inflammatory genes Il-6 and Il-13, were downregulated after an infection caused by the 267/47 strain compared to those after infection with the H37Rv strain.
Collapse
Affiliation(s)
- Mikhail V. Fursov
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Egor A. Shitikov
- Federal Research and Clinical Center of Physical-Chemical Medicine, 119435 Moscow, Russia; (E.A.S.); (D.A.B.)
| | - Denis A. Lagutkin
- National Medical Research Center for Phthisiopulmonology and Infectious Diseases of the Ministry of Health of the Russian Federation, 127473 Moscow, Russia; (D.A.L.); (A.E.P.); (A.S.V.); (I.A.V.)
| | - Anastasiia D. Fursova
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Elena A. Ganina
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Tatiana I. Kombarova
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Natalia S. Grishenko
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Tatiana I. Rudnitskaya
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Dmitry A. Bespiatykh
- Federal Research and Clinical Center of Physical-Chemical Medicine, 119435 Moscow, Russia; (E.A.S.); (D.A.B.)
| | - Nadezhda V. Kolupaeva
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Viktoria V. Firstova
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Lubov V. Domotenko
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Anna E. Panova
- National Medical Research Center for Phthisiopulmonology and Infectious Diseases of the Ministry of Health of the Russian Federation, 127473 Moscow, Russia; (D.A.L.); (A.E.P.); (A.S.V.); (I.A.V.)
| | - Anatoliy S. Vinokurov
- National Medical Research Center for Phthisiopulmonology and Infectious Diseases of the Ministry of Health of the Russian Federation, 127473 Moscow, Russia; (D.A.L.); (A.E.P.); (A.S.V.); (I.A.V.)
| | - Vladimir A. Gushchin
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (V.A.G.); (A.P.T.)
| | - Artem P. Tkachuk
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (V.A.G.); (A.P.T.)
| | - Irina A. Vasilyeva
- National Medical Research Center for Phthisiopulmonology and Infectious Diseases of the Ministry of Health of the Russian Federation, 127473 Moscow, Russia; (D.A.L.); (A.E.P.); (A.S.V.); (I.A.V.)
| | - Vasiliy D. Potapov
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| | - Ivan A. Dyatlov
- State Research Center for Applied Microbiology and Biotechnology, Territory “Kvartal A”, 142279 Serpukhov, Russia; (A.D.F.); (E.A.G.); (T.I.K.); (N.S.G.); (T.I.R.); (N.V.K.); (V.V.F.); (L.V.D.); (V.D.P.); (I.A.D.)
| |
Collapse
|
43
|
Cortés HD, Gómez FA, Marshall SH. The Phagosome-Lysosome Fusion Is the Target of a Purified Quillaja saponin Extract (PQSE) in Reducing Infection of Fish Macrophages by the Bacterial Pathogen Piscirickettsia salmonis. Antibiotics (Basel) 2021; 10:antibiotics10070847. [PMID: 34356768 PMCID: PMC8300623 DOI: 10.3390/antibiotics10070847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/20/2022] Open
Abstract
Piscirickettsia salmonis, the etiological agent of Piscirickettsiosis, is a Gram-negative and facultative intracellular pathogen that has affected the Chilean salmon industry since 1989. The bacterium is highly aggressive and can survive and replicate within fish macrophages using the Dot/Icm secretion system to evade the host’s immune response and spread systemically. To date, no efficient control measures have been developed for this disease; therefore, the producers use large amounts of antibiotics to control this pathogen. In this frame, this work has focused on evaluating the use of saponins from Quillaja saponaria as a new alternative to control the Piscirickettsiosis. It has been previously reported that purified extract of Q. saponaria (PQSE) displays both antimicrobial activity against pathogenic bacteria and viruses and adjuvant properties. Our results show that PQSE does not present antimicrobial activity against P. salmonis, although it reduces P. salmonis infection in an in vitro model, promoting the phagosome–lysosome fusion. Additionally, we demonstrate that PQSE modulates the expression of IL-12 and IL-10 in infected cells, promoting the immune response against the pathogen and reducing the expression of pathogen virulence genes. These results together strongly argue for specific anti-invasion and anti-intracellular replication effects induced by the PQSE in macrophages.
Collapse
|
44
|
Jiang H, Cui B, Zhang J. Mycobacterium tuberculosis (MTB) antigen-induced upregulation of interleukin-35 expression in patients with MTB infection: In vitro blockade of the effects of interleukin-35 on T lymphocyte subsets. Pathog Dis 2021; 79:6318859. [PMID: 34245560 DOI: 10.1093/femspd/ftab035] [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: 04/19/2021] [Accepted: 07/08/2021] [Indexed: 11/14/2022] Open
Abstract
Immunosuppressive interleukin-35 (IL-35) serum concentrations were analyzed in patients with active pulmonary Mycobacterium tuberculosis (MTB) infections (PTB), PTB patients after two months treatment (stable PTB) and healthy controls. IL-35 concentrations were highest in active PTB followed by stable PTB cases and lowest in healthy control participants (all P < 0.01). The same trents were found for supernatants of isolated blood mononuclear cells (PBMCs), with additional enhancements after MTB antigen stimulation only for PBMCs of active and stable PTB patients (P < 0.001), for EBI3 and IL-12a transcriptions in PBMCs (P < 0.001) and percentages of EBI3 expressing (CD4 + CD25 + Foxp3+) regulatory T cells (Treg) (P < 0.001). IL-35 antibody applications significantly reversed MTB antigen stimulated IL-35 and IL-10 expression in PBMCs of active and stable PTB patients, and reduced Foxp3 expression in CD4 + CD25 + cells and EBI3 expression in Treg cells, but had no effects on healthy control cells. The percentages of Th1 and Th17 cells in CD4 + cells were enhanced after MTB antigen stimulation of cells taken from active and stable PTB patients, which were partly increased only for Th1 cells after IL-35 antibody exposure. MTB antigen-driven upregulation of IL-35 may lead to reduced immune surveillance in PTB patients.
Collapse
Affiliation(s)
- Hongbin Jiang
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Yangpu District, Shanghai 200433, China
| | - Beinian Cui
- Department of Laboratory Medicine, Shanghai DeltaHealth Hospital. No. 109 Xule Road, Xujing Town, Qingpu District, Shanghai 201702, China
| | - Jun Zhang
- Department of Laboratory Medicine, Shanghai DeltaHealth Hospital. No. 109 Xule Road, Xujing Town, Qingpu District, Shanghai 201702, China
| |
Collapse
|
45
|
A Small Protein but with Diverse Roles: A Review of EsxA in Mycobacterium-Host Interaction. Cells 2021; 10:cells10071645. [PMID: 34209120 PMCID: PMC8305481 DOI: 10.3390/cells10071645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/20/2022] Open
Abstract
As a major effector of the ESX-1 secretion system, EsxA is essential for the virulence of pathogenic mycobacteria, such as Mycobacterium tuberculosis (Mtb) and Mycobacterium marinum (Mm). EsxA possesses an acidic pH-dependent membrane permeabilizing activity and plays an essential role by mediating mycobacterial escape from the phagosome and translocation to the cytosol for intracellular replication. Moreover, EsxA regulates host immune responses as a potent T-cell antigen and a strong immunoregulator. EsxA interacts with multiple cellular proteins and stimulates several signal pathways, such as necrosis, apoptosis, autophagy, and antigen presentation. Interestingly, there is a co-dependency in the expression and secretion of EsxA and other mycobacterial factors, which greatly increases the complexity of dissecting the precise roles of EsxA and other factors in mycobacterium-host interaction. In this review, we summarize the current understandings of the roles and functions of EsxA in mycobacterial infection and discuss the challenges and future directions.
Collapse
|
46
|
Park HS, Back YW, Jang IT, Lee KI, Son YJ, Choi HG, Dang TB, Kim HJ. Mycobacterium tuberculosis Rv2145c Promotes Intracellular Survival by STAT3 and IL-10 Receptor Signaling. Front Immunol 2021; 12:666293. [PMID: 34017340 PMCID: PMC8129509 DOI: 10.3389/fimmu.2021.666293] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
Although Mycobacterium tuberculosis (Mtb) is an intracellular pathogen in phagocytic cells, the factors and mechanisms by which they invade and persist in host cells are still not well understood. Characterization of the bacterial proteins modulating macrophage function is essential for understanding tuberculosis pathogenesis and bacterial virulence. Here we investigated the pathogenic role of the Rv2145c protein in stimulating IL-10 production. We first found that recombinant Rv2145c stimulated bone marrow-derived macrophages (BMDMs) to secrete IL-10, IL-6 and TNF-α but not IL-12p70 and to increase the expression of surface molecules through the MAPK, NF-κB, and TLR4 pathways and enhanced STAT3 activation and the expression of IL-10 receptor in Mtb-infected BMDMs. Rv2145c significantly enhanced intracellular Mtb growth in BMDMs compared with that in untreated cells, which was abrogated by STAT3 inhibition and IL-10 receptor (IL-10R) blockade. Expression of Rv2145c in Mycobacterium smegmatis (M. smegmatis) led to STAT3-dependent IL-10 production and enhancement of intracellular growth in BMDMs. Furthermore, the clearance of Rv2145c-expressing M. smegmatis in the lungs and spleens of mice was delayed, and these effects were abrogated by administration of anti-IL-10R antibodies. Finally, all mice infected with Rv2145c-expressing M. smegmatis died, but those infected with the vector control strain did not. Our data suggest that Rv2145c plays a role in creating a favorable environment for bacterial survival by modulating host signals.
Collapse
Affiliation(s)
- Hye-Soo Park
- Department of Microbiology and Medical Science, and Translational Immunology Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Yong Woo Back
- Department of Microbiology and Medical Science, and Translational Immunology Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - In-Taek Jang
- Department of Microbiology and Medical Science, and Translational Immunology Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Kang-In Lee
- Department of Microbiology and Medical Science, and Translational Immunology Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Yeo-Jin Son
- Department of Microbiology and Medical Science, and Translational Immunology Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Han-Gyu Choi
- Department of Microbiology and Medical Science, and Translational Immunology Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Thi Binh Dang
- Department of Microbiology and Medical Science, and Translational Immunology Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology and Medical Science, and Translational Immunology Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
47
|
Etna MP, Severa M, Licursi V, Pardini M, Cruciani M, Rizzo F, Giacomini E, Macchia G, Palumbo O, Stallone R, Carella M, Livingstone M, Negri R, Pellegrini S, Coccia EM. Genome-Wide Gene Expression Analysis of Mtb-Infected DC Highlights the Rapamycin-Driven Modulation of Regulatory Cytokines via the mTOR/GSK-3β Axis. Front Immunol 2021; 12:649475. [PMID: 33936070 PMCID: PMC8086600 DOI: 10.3389/fimmu.2021.649475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
In human primary dendritic cells (DC) rapamycin-an autophagy inducer and protein synthesis inhibitor-overcomes the autophagy block induced by Mycobacterium tuberculosis (Mtb) and promotes a Th1 response via IL-12 secretion. Here, the immunostimulatory activity of rapamycin in Mtb-infected DC was further investigated by analyzing both transcriptome and translatome gene profiles. Hundreds of differentially expressed genes (DEGs) were identified by transcriptome and translatome analyses of Mtb-infected DC, and some of these genes were found further modulated by rapamycin. The majority of transcriptome-associated DEGs overlapped with those present in the translatome, suggesting that transcriptionally stimulated mRNAs are also actively translated. In silico analysis of DEGs revealed significant changes in intracellular cascades related to cytokine production, cytokine-induced signaling and immune response to pathogens. In particular, rapamycin treatment of Mtb-infected DC caused an enrichment of IFN-β, IFN-λ and IFN-stimulated gene transcripts in the polysome-associated RNA fraction. In addition, rapamycin led to an increase of IL-12, IL-23, IL-1β, IL-6, and TNF-α but to a reduction of IL-10. Interestingly, upon silencing or pharmacological inhibition of GSK-3β, the rapamycin-driven modulation of the pro- and anti-inflammatory cytokine balance was lost, indicating that, in Mtb-infected DC, GSK-3β acts as molecular switch for the regulation of the cytokine milieu. In conclusion, our study sheds light on the molecular mechanism by which autophagy induction contributes to DC activation during Mtb infection and points to rapamycin and GSK-3β modulators as promising compounds for host-directed therapy in the control of Mtb infection.
Collapse
Affiliation(s)
- Marilena P Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology, Sapienza University, Rome, Italy
| | - Manuela Pardini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Melania Cruciani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Raffaella Stallone
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Massimo Carella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Mark Livingstone
- Cytokine Signaling Unit, Inserm, Institut Pasteur, Paris, France
| | - Rodolfo Negri
- Department of Biology and Biotechnology, Sapienza University, Rome, Italy
| | | | - Eliana M Coccia
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
48
|
Ó Maoldomhnaigh C, Cox DJ, Phelan JJ, Malone FD, Keane J, Basdeo SA. The Warburg Effect Occurs Rapidly in Stimulated Human Adult but Not Umbilical Cord Blood Derived Macrophages. Front Immunol 2021; 12:657261. [PMID: 33927724 PMCID: PMC8076563 DOI: 10.3389/fimmu.2021.657261] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
The Warburg effect, defined as increased glycolysis and decreased oxidative phosphorylation, occurs in murine macrophages following LPS stimulation and is required for activation. There are differences between human and murine macrophage metabolic responses to stimulation, with peak metabolite concentrations occurring earlier in humans than mice. Complex changes occur in the human immune system with age, resulting in the very young and the very old being more susceptible to infections. Anti-bacterial immune responses in umbilical cord immune cells are considered deficient but there is a paucity of data on the role that metabolism plays. We hypothesized that metabolic responses in human macrophages occur early during activation. In addition, we hypothesized that umbilical cord derived macrophages have an altered immunometabolic response compared with adult macrophages. We demonstrate that adult and cord blood monocyte derived macrophages (MDM) immediately increase glycolysis in response to stimulation with LPS or Mycobacterium tuberculosis (Mtb), however only adult MDM decrease oxidative phosphorylation. At 24 hours post stimulation, glycolysis remains elevated in both adult and cord blood MDM, oxidative phosphorylation remains unchanged in the cord blood MDM and has normalized in the adult MDM stimulated with Mtb. However, LPS stimulated adult MDM have increased oxidative phosphorylation at 24 hours, illustrating differences in metabolic responses to different stimuli, time-dependent variation in responses and differences in macrophage metabolism in adults compared with umbilical cord blood. We compared the phenotype and function of macrophages derived from adult or cord blood. Cord blood MDM secreted less TNF following Mtb stimulation and more IL-6 following LPS stimulation compared with adult MDM. Our findings demonstrate that whilst cord blood MDM exhibit an immediate increase in glycolytic flux in response to stimulation, similar to adult MDM, cord blood MDM do not concomitantly decrease oxygen consumption. This indicates that adult macrophages shift to Warburg metabolism immediately after stimulation, but cord blood macrophages do not. Understanding the differences in the metabolic profiles of macrophages over a human lifetime will enable the translation of immunometabolism into effective immuno-supportive therapies that could potentially be targeted at vulnerable populations, such as the very old and the very young.
Collapse
Affiliation(s)
- Cilian Ó Maoldomhnaigh
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Donal J Cox
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - James J Phelan
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Fergal D Malone
- Obstetrics & Gynecology, Royal College of Surgeons in Ireland, Rotunda Hospital, Dublin, Ireland
| | - Joseph Keane
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Sharee A Basdeo
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| |
Collapse
|
49
|
Bortell N, Aguilera ER, Lenz LL. Pulmonary insults exacerbate susceptibility to oral Listeria monocytogenes infection through the production of IL-10 by NK cells. PLoS Pathog 2021; 17:e1009531. [PMID: 33878120 PMCID: PMC8087096 DOI: 10.1371/journal.ppat.1009531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/30/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022] Open
Abstract
Most individuals who consume foods contaminated with the bacterial pathogen Listeria monocytogenes (Lm) develop mild symptoms, while others are susceptible to life-threatening systemic infections (listeriosis). Although it is known that the risk of severe disease is increased in certain human populations, including the elderly, it remains unclear why others who consume contaminated food develop listeriosis. Here, we used a murine model to discover that pulmonary coinfections can impair the host's ability to adequately control and eradicate systemic Lm that cross from the intestines to the bloodstream. We found that the resistance of mice to oral Lm infection was dramatically reduced by coinfection with Streptococcus pneumoniae (Spn), a bacterium that colonizes the respiratory tract and can also cause severe infections in the elderly. Exposure to Spn or microbial products, including a recombinant Lm protein (L1S) and lipopolysaccharide (LPS), rendered otherwise resistant hosts susceptible to severe systemic Lm infection. In addition, we show that this increase in susceptibility was dependent on an increase in the production of interleukin-10 (IL-10) from Ncr1+ cells, including natural killer (NK) cells. Lastly, the ability of Ncr1+ cell derived IL-10 to increase disease susceptibility correlated with a dampening of both myeloid cell accumulation and myeloid cell phagocytic capacity in infected tissues. These data suggest that efforts to minimize inflammation in response to an insult at the respiratory mucosa render the host more susceptible to infections by Lm and possibly other pathogens that access the oral mucosa.
Collapse
Affiliation(s)
- Nikki Bortell
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Elizabeth R. Aguilera
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Laurel L. Lenz
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| |
Collapse
|
50
|
Mawa PA, Hasso-Agopsowicz M, Lubyayi L, Nabakooza G, Nakibuule M, Blitz R, Dun L, Govind A, Kaleebu P, Webb EL, Elliott AM, Dockrell HM, Cose S, Smith SG. Immune Responses Following BCG Immunization of Infants in Uganda and United Kingdom Are Similar for Purified Protein Derivative but Differ for Secretory Proteins of Mycobacterium tuberculosis. Front Immunol 2021; 12:637114. [PMID: 33815390 PMCID: PMC8017231 DOI: 10.3389/fimmu.2021.637114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/22/2021] [Indexed: 11/15/2022] Open
Abstract
Introduction: The immunogenicity of BCG vaccination in infants differs between populations. We hypothesized that prenatal exposure to mycobacterial antigens might explain the differences in immune responses to BCG seen in other studies of infants in Africa and the United Kingdom (UK) and we explored this in birth cohorts in Uganda and the UK. Materials and Methods: Blood samples were obtained from BCG-immunized infants of mothers with (n = 110) and without (n = 121) latent Mycobacterium tuberculosis infection (LTBI) in Uganda and BCG-immunized infants of mothers without LTBI (n = 25) in the UK at 10 and 52 weeks after birth. Cytokine and chemokine responses to PPD were measured to assess responses to BCG immunization, and to ESAT6/CFP10 to assess exposure to or infection with M. tuberculosis or non-tuberculous mycobacteria (NTM) in 6-day whole blood culture supernatants by a 17-plex Luminex assay. Median responses were compared between Ugandan infants (together, and separated by maternal LTBI status) and UK infants. Results: The IFN-γ response to BCG vaccination was similar between Ugandan and UK infants at 10 and 52 weeks. At week 52, TNF production was marginally higher in Ugandan infants, but after adjusting for multiple comparisons this difference was not significant. At weeks 10 and 52, stimulation of blood with ESAT6/CFP10 produced significantly higher IFN-γ, TNF, IL-12p40, IL-1α, IL-1β, IL-1Ra, IP-10, MIP-1α, MIP-1β, and GM-CSF in Ugandan compared to UK infants. Stimulation of blood with ESAT6/CFP10 produced significantly higher amounts of IL-8 (p = 0.0001), IL-10 (p = 0.0022), and IL-13 (p = 0.0020) in the UK than in Ugandan infants of mothers without LTBI at week 10, but not at week 52. Conclusions: Immune responses to mycobacterial antigens following BCG immunization are similar for PPD, but differ for ESAT6/CFP10, between infants in Uganda and the UK. Neither maternal LTBI nor infant exposure to or infection with mycobacteria impacts the response to BCG. The observed global differences in immune response to BCG immunization are likely to be due to other causes.
Collapse
Affiliation(s)
- Patrice A. Mawa
- Immunomodulation and Vaccines Programme, Medical Research Council-Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Mateusz Hasso-Agopsowicz
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Lawrence Lubyayi
- Immunomodulation and Vaccines Programme, Medical Research Council-Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Epidemiology and Biostatistics, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Grace Nabakooza
- Immunomodulation and Vaccines Programme, Medical Research Council-Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Marjorie Nakibuule
- Immunomodulation and Vaccines Programme, Medical Research Council-Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Rose Blitz
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Li Dun
- Fetal Medicine Unit, Gynaecology and Obstetrics Department, North Middlesex University Hospital National Health Service Trust, London, United Kingdom
| | - Abha Govind
- Fetal Medicine Unit, Gynaecology and Obstetrics Department, North Middlesex University Hospital National Health Service Trust, London, United Kingdom
| | - Pontiano Kaleebu
- Immunomodulation and Vaccines Programme, Medical Research Council-Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Emily L. Webb
- Medical Research Council Tropical Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alison M. Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council-Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Hazel M. Dockrell
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Stephen Cose
- Immunomodulation and Vaccines Programme, Medical Research Council-Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Steven G. Smith
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|