1
|
Baya B, Diarra B, Dabitao DK, Somboro A, Traore FG, Goita D, Coulibaly G, Sanogo M, Wague M, Kone B, Kone D, Ouattara K, Soumare D, Kanoute T, Toloba Y, Maiga AI, Maiga M, Diallo S, Murphy RL, Doumbia S. High Schistosoma mansoni Co-Infection in Tuberculosis Patients with or without Human Immunodeficiency Virus: A Prospective Cohort Study. RESEARCH SQUARE 2024:rs.3.rs-4796634. [PMID: 39281870 PMCID: PMC11398560 DOI: 10.21203/rs.3.rs-4796634/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background People with Latent tuberculosis infection (LTBI) remain the reservoir of tuberculosis. One-third to 1/4 of the world's population is infected. Its reactivation is due to factors that disrupt the host's immune response. Recent findings showed that Schistosoma mansoni coinfection leads to a Th2/Th1 profile which results in an immune modulation that favors the escape of the Mycobacteria. Schistosoma mansoni may contribute to TB incidence in endemic regions. We aimed to investigate the co-infection rate and patient outcomes. Methods A prospective cohort study was conducted between 2020-2022 at University Clinical Research Center (UCRC), including culture-confirmed active pulmonary TB patients and tested for Schistosoma mansoni in stools using Kato-Katz Technique. After descriptive analysis a logistic regression was performed to determine risk factors associated with TB and Schistosoma mansoni co-infection. Results Data of 174 tuberculosis-confirmed patients, Kato-Katz tested were analyzed. Males represented 62.6%, mean age was 34.9 ± 13.8 years, 29.9% were smokers, alcohol consumption 13.8%, TB contact history 26.4%, HIV coinfection 11.5%, diabetes 6.3%, undernourished 55.7%. Schistosoma mansoni prevalence was 28.7%. The co-infection was associated with less lung cavitation [aOR = 0.24 [95% CI (0.06-0.85), p = 0.028], unfavorable treatment result [aOR = 2.95 (1.23-7.08), p = 0.015] and death [aOR = 3.43 (1.12-10.58), p = 0.032]. Conclusions Despite Kato-Katz's low sensitivity, Schistosoma mansoni coinfection was found in one-third of the TB patients; 2.5-fold higher than that of HIV. The coinfection was associated with poor treatment results and death.
Collapse
Affiliation(s)
- Bocar Baya
- University Clinical Research Center (UCRC)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Kaushal D, Sharan R, Zou Y, Lai Z, Singh B, Shivanna V, Dick E, Hall-Ursone S, Khader S, Mehra S, Alvarez X, Rengarajan J. Concurrent TB and HIV therapies effectively control clinical reactivation of TB during co-infection but fail to eliminate chronic immune activation. RESEARCH SQUARE 2024:rs.3.rs-4908400. [PMID: 39257997 PMCID: PMC11384027 DOI: 10.21203/rs.3.rs-4908400/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The majority of Human Immunodeficiency Virus (HIV) negative individuals exposed to Mycobacterium tuberculosis (Mtb) control the bacillary infection as latent TB infection (LTBI). Co-infection with HIV, however, drastically increases the risk to progression to tuberculosis (TB) disease. TB is therefore the leading cause of death in people living with HIV (PLWH) globally. Combinatorial antiretroviral therapy (cART) is the cornerstone of HIV care in humans and reduces the risk of reactivation of LTBI. However, the immune control of Mtb infection is not fully restored by cART as indicated by higher incidence of TB in PLWH despite cART. In the macaque model of co-infection, skewed pulmonary CD4+ TEM responses persist, and new TB lesions form despite cART treatment. We hypothesized that regimens that concurrently administer anti-TB therapy and cART would significantly reduce TB in co-infected macaques than cART alone, resulting in superior bacterial control, mitigation of persistent inflammation and lasting protective immunity. We studied components of TB immunity that remain impaired after cART in the lung compartment, versus those that are restored by concurrent 3 months of once weekly isoniazid and rifapentine (3HP) and cART in the rhesus macaque (RM) model of LTBI and Simian Immunodeficiency Virus (SIV) co-infection. Concurrent administration of cART + 3HP did improve clinical and microbiological attributes of Mtb/SIV co-infection compared to cART-naïve or -untreated RMs. While RMs in the cART + 3HP group exhibited significantly lower granuloma volumes after treatment, they, however, continued to harbor caseous granulomas with increased FDG uptake. cART only partially restores the constitution of CD4 + T cells to the lung compartment in co-infected macaques. Concurrent therapy did not further enhance the frequency of reconstituted CD4+ T cells in BAL and lung of Mtb/SIV co-infected RMs compared to cART, and treated animals continued to display incomplete reconstitution to the lung. Furthermore, the reconstituted CD4+ T cells in BAL and lung of cART + 3HP treated RMs exhibited an increased frequencies of activated, exhausted and inflamed phenotype compared to LTBI RMs. cART + 3HP failed to restore the effector memory CD4+ T cell population that was significantly reduced in pulmonary compartment post SIV co-infection. Concurrent therapy was associated with the induction of Type I IFN transcriptional signatures and led to increased Mtb-specific TH1/TH17 responses correlated with protection, but decreased Mtb-specific TNFa responses, which could have a detrimental impact on long term protection. Our results suggest the mechanisms by which Mtb/HIV co-infected individuals remain at risk for progression due to subsequent infections or reactivation due of persisting defects in pulmonary T cell responses. By identifying lung-specific immune components in this model, it is possible to pinpoint the pathways that can be targeted for host-directed adjunctive therapies for TB/HIV co-infection.
Collapse
Affiliation(s)
- Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute
| | | | | | - Zhao Lai
- The University of Texas Health San Antonio
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Huang S, Liu M, Zhang H, Song W, Guo W, Feng Y, Ma X, Shi X, Liu J, Liu L, Qi T, Wang Z, Yan B, Shen Y. HIV-MTB Co-Infection Reduces CD4+ T Cells and Affects Granuloma Integrity. Viruses 2024; 16:1335. [PMID: 39205309 PMCID: PMC11360352 DOI: 10.3390/v16081335] [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: 06/22/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Granuloma is a crucial pathological feature of tuberculosis (TB). The relationship between CD4+ T cells in both peripheral blood and granulomatous tissue, and the integrity of granulomas in Human Immunodeficiency Virus (HIV)-MTB co-infection, remains unexplored. This study collected biopsy specimens from 102 TB patients (53 with HIV-MTB co-infection and 49 only with TB). Hematoxylin and eosin (HE) staining and immunohistochemical staining were performed, followed by microscopic examination of the integrity of tuberculous granulomas. Through statistical analysis of peripheral blood CD4+ T cell counts, tissue CD4+ T cell proportion, and the integrity of granulomas, it was observed that HIV infection leads to poor formation of tuberculous granulomas. Peripheral blood CD4+ T cell counts were positively correlated with granuloma integrity, and there was a similar positive correlation between tissue CD4+ T cell proportions and granuloma integrity. Additionally, a positive correlation was found between peripheral blood CD4+ T cell counts and the proportion of CD4+ T cells in granuloma tissues. Therefore, HIV infection could impact the morphology and structure of tuberculous granulomas, with a reduced proportion of both peripheral blood and tissue CD4+ T lymphocytes.
Collapse
Affiliation(s)
- Suyue Huang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Maoying Liu
- Department of Microbiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Hui Zhang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Song
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Wenjuan Guo
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Yanling Feng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Xin Ma
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Xia Shi
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Jianjian Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Li Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Tangkai Qi
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Zhenyan Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Bo Yan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Yinzhong Shen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| |
Collapse
|
4
|
Anes E, Azevedo-Pereira JM, Pires D. Role of Type I Interferons during Mycobacterium tuberculosis and HIV Infections. Biomolecules 2024; 14:848. [PMID: 39062562 PMCID: PMC11275242 DOI: 10.3390/biom14070848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Tuberculosis and AIDS remain two of the most relevant human infectious diseases. The pathogens that cause them, Mycobacterium tuberculosis (Mtb) and HIV, individually elicit an immune response that treads the line between beneficial and detrimental to the host. Co-infection further complexifies this response since the different cytokines acting on one infection might facilitate the dissemination of the other. In these responses, the role of type I interferons is often associated with antiviral mechanisms, while for bacteria such as Mtb, their importance and clinical relevance as a suitable target for manipulation are more controversial. In this article, we review the recent knowledge on how these interferons play distinct roles and sometimes have opposite consequences depending on the stage of the pathogenesis. We highlight the dichotomy between the acute and chronic infections displayed by both infections and how type I interferons contribute to an initial control of each infection individually, while their chronic induction, particularly during HIV infection, might facilitate Mtb primo-infection and progression to disease. We expect that further findings and their systematization will allow the definition of windows of opportunity for interferon manipulation according to the stage of infection, contributing to pathogen clearance and control of immunopathology.
Collapse
Affiliation(s)
- Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
| | - David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Estrada Octávio Pato, 2635-631 Rio de Mouro, Portugal
| |
Collapse
|
5
|
Singh B, Sharan R, Ravichandran G, Escobedo R, Shivanna V, Dick EJ, Hall-Ursone S, Arora G, Alvarez X, Singh DK, Kaushal D, Mehra S. Indoleamine-2,3-dioxygenase inhibition improves immunity and is safe for concurrent use with cART during Mtb/SIV coinfection. JCI Insight 2024; 9:e179317. [PMID: 39114981 PMCID: PMC11383603 DOI: 10.1172/jci.insight.179317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/26/2024] [Indexed: 09/13/2024] Open
Abstract
Chronic immune activation promotes tuberculosis (TB) reactivation in the macaque Mycobacterium tuberculosis (M. tuberculosis)/SIV coinfection model. Initiating combinatorial antiretroviral therapy (cART) early lowers the risk of TB reactivation, but immune activation persists. Studies of host-directed therapeutics (HDTs) that mitigate immune activation are, therefore, required. Indoleamine 2,3, dioxygenase (IDO), a potent immunosuppressor, is one of the most abundantly induced proteins in NHP and human TB granulomas. Inhibition of IDO improves immune responses in the lung, leading to better control of TB, including adjunctive to TB chemotherapy. The IDO inhibitor D-1 methyl tryptophan (D1MT) is, therefore, a bona fide TB HDT candidate. Since HDTs against TB are likely to be deployed in an HIV coinfection setting, we studied the effect of IDO inhibition in M. tuberculosis/SIV coinfection, adjunctive to cART. D1MT is safe in this setting, does not interfere with viral suppression, and improves the quality of CD4+ and CD8+ T cell responses, including reconstitution, activation and M. tuberculosis-specific cytokine production, and access of CD8+ T cells to the lung granulomas; it reduces granuloma size and necrosis, type I IFN expression, and the recruitment of inflammatory IDO+ interstitial macrophages (IMs). Thus, trials evaluating the potential of IDO inhibition as HDT in the setting of cART in M. tuberculosis/HIV coinfected individuals are warranted.
Collapse
|
6
|
Vyklyuk Y, Semianiv I, Nevinskyi D, Todoriko L, Boyko N. Applying geospatial multi-agent system to model various aspects of tuberculosis transmission. New Microbes New Infect 2024; 59:101417. [PMID: 38737327 PMCID: PMC11088189 DOI: 10.1016/j.nmni.2024.101417] [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: 01/13/2024] [Revised: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction The paper presents epidemiological process modeling, with a focus on tuberculosis utilizing multi-agent system. Material and methods This study involves the development of an algorithm that harnesses the potential of artificial intelligence to create a geospatial model that highlights the different pathways of TB transmission. The modeling process itself is characterized by a series of key stages, including initialization of the city, calibration of health parameters, simulation of the working day, propagation of the spread of infection, the evolution of disease trajectories, rigorous statistical calculations and transition to the following day. A comprehensive description of the course of active tuberculosis is presented, following the official hypothesis recommended by the World Health Organization. A comprehensive simulation, illustrating the propagation of tuberculosis in an entirely healthy environment devoid of any preventive or therapeutic measures, is presented. To ascertain the adequacy of the model and its sensitivity to the principal parameters governing the course of tuberculosis, a series of experiments were meticulously conducted, employing three distinct approximations, namely: the basic model, the model incorporating mortality factors, and the comprehensive model, encompassing all relevant aspects. Conclusions The model's results exhibit stability and lack of significant fluctuations. The statistical values obtained for infected, latent, and recovered individuals align well with known medical data, confirming the model's adequacy.
Collapse
Affiliation(s)
- Yaroslav Vyklyuk
- Lviv Polytechnic National University, The Department of Artificial Intelligence, Lviv, Ukraine
| | - Ihor Semianiv
- Bukovinian State Medical University, The Department of Pathobiology and Pulmonology, Chernivtsi, Ukraine
| | - Denys Nevinskyi
- Lviv Polytechnic National University, The Department of Electronics and Information Technology, Lviv, Ukraine
| | - Lilia Todoriko
- Bukovinian State Medical University, The Department of Pathobiology and Pulmonology, Chernivtsi, Ukraine
| | - Nataliya Boyko
- Lviv Polytechnic National University, The Department of Artificial Intelligence, Lviv, Ukraine
| |
Collapse
|
7
|
Gyu Choi H, Woong Kwon K, Jae Shin S. Importance of adjuvant selection in tuberculosis vaccine development: Exploring basic mechanisms and clinical implications. Vaccine X 2023; 15:100400. [PMID: 37965276 PMCID: PMC10641539 DOI: 10.1016/j.jvacx.2023.100400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
The global emergency of unexpected pathogens, exemplified by SARS-CoV-2, has emphasized the importance of vaccines in thwarting infection and curtailing the progression of severe disease. The scourge of tuberculosis (TB), emanating from the Mycobacterium tuberculosis (Mtb) complex, has inflicted a more profound toll in terms of mortality and morbidity than any other infectious agents prior to the SARS-CoV-2 pandemic. Despite the existence of Bacillus Calmette-Guérin (BCG), the only licensed vaccine developed a century ago, its efficacy against TB remains unsatisfactory, particularly in preventing pulmonary Mtb infections in adolescents and adults. However, collaborations between academic and industrial entities have led to a renewed impetus in the development of TB vaccines, with numerous candidates, particularly subunit vaccines with specialized adjuvants, exhibiting promising outcomes in recent clinical studies. Adjuvants are crucial in modulating optimal immunological responses, by endowing immune cells with sufficient antigen and immune signals. As exemplified by the COVID-19 vaccine landscape, the interplay between vaccine efficacy and adverse effects is of paramount importance, particularly for the elderly and individuals with underlying ailments such as diabetes and concurrent infections. In this regard, adjuvants hold the key to optimizing vaccine efficacy and safety. This review accentuates the pivotal roles of adjuvants and their underlying mechanisms in the development of TB vaccines. Furthermore, we expound on the prospects for the development of more efficacious adjuvants and their synergistic combinations for individuals in diverse states, such as aging, HIV co-infection, and diabetes, by examining the immunological alterations that arise with aging and comparing them with those observed in younger cohorts.
Collapse
Affiliation(s)
- Han Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
8
|
Sankar P, Mishra BB. Early innate cell interactions with Mycobacterium tuberculosis in protection and pathology of tuberculosis. Front Immunol 2023; 14:1260859. [PMID: 37965344 PMCID: PMC10641450 DOI: 10.3389/fimmu.2023.1260859] [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: 07/18/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
Tuberculosis (TB) remains a significant global health challenge, claiming the lives of up to 1.5 million individuals annually. TB is caused by the human pathogen Mycobacterium tuberculosis (Mtb), which primarily infects innate immune cells in the lungs. These immune cells play a critical role in the host defense against Mtb infection, influencing the inflammatory environment in the lungs, and facilitating the development of adaptive immunity. However, Mtb exploits and manipulates innate immune cells, using them as favorable niche for replication. Unfortunately, our understanding of the early interactions between Mtb and innate effector cells remains limited. This review underscores the interactions between Mtb and various innate immune cells, such as macrophages, dendritic cells, granulocytes, NK cells, innate lymphocytes-iNKT and ILCs. In addition, the contribution of alveolar epithelial cell and endothelial cells that constitutes the mucosal barrier in TB immunity will be discussed. Gaining insights into the early cellular basis of immune reactions to Mtb infection is crucial for our understanding of Mtb resistance and disease tolerance mechanisms. We argue that a better understanding of the early host-pathogen interactions could inform on future vaccination approaches and devise intervention strategies.
Collapse
Affiliation(s)
| | - Bibhuti Bhusan Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| |
Collapse
|
9
|
Kulkarni S, Endsley JJ, Lai Z, Bradley T, Sharan R. Single-Cell Transcriptomics of Mtb/HIV Co-Infection. Cells 2023; 12:2295. [PMID: 37759517 PMCID: PMC10529032 DOI: 10.3390/cells12182295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/17/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Tuberculosis (TB) and Human Immunodeficiency Virus (HIV) co-infection continues to pose a significant healthcare burden. HIV co-infection during TB predisposes the host to the reactivation of latent TB infection (LTBI), worsening disease conditions and mortality. There is a lack of biomarkers of LTBI reactivation and/or immune-related transcriptional signatures to distinguish active TB from LTBI and predict TB reactivation upon HIV co-infection. Characterizing individual cells using next-generation sequencing-based technologies has facilitated novel biological discoveries about infectious diseases, including TB and HIV pathogenesis. Compared to the more conventional sequencing techniques that provide a bulk assessment, single-cell RNA sequencing (scRNA-seq) can reveal complex and new cell types and identify more high-resolution cellular heterogeneity. This review will summarize the progress made in defining the immune atlas of TB and HIV infections using scRNA-seq, including host-pathogen interactions, heterogeneity in HIV pathogenesis, and the animal models employed to model disease. This review will also address the tools needed to bridge the gap between disease outcomes in single infection vs. co-infection. Finally, it will elaborate on the translational benefits of single-cell sequencing in TB/HIV diagnosis in humans.
Collapse
Affiliation(s)
- Smita Kulkarni
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Janice J. Endsley
- Departments of Microbiology & Immunology and Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, The University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| | - Todd Bradley
- Genomic Medicine Center, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, MO 64108, USA;
- Departments of Pediatrics and Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO 66160, USA
- Department of Pediatrics, UMKC School of Medicine, Kansas City, MO 64108, USA
| | - Riti Sharan
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| |
Collapse
|
10
|
Kaushal D, Singh DK, Mehra S. Immune Responses in Lung Granulomas during Mtb/HIV Co-Infection: Implications for Pathogenesis and Therapy. Pathogens 2023; 12:1120. [PMID: 37764928 PMCID: PMC10534770 DOI: 10.3390/pathogens12091120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
HIV and TB are the cause of significant worldwide mortality and pose a grave danger to the global public health. TB is the leading cause of death in HIV-infected persons, with one in four deaths attributable to TB. While the majority of healthy individuals infected with M. tuberculosis (Mtb) are able to control the infection, co-infection with HIV increases the risk of TB infection progressing to TB disease by over 20-fold. While antiretroviral therapy (ART), the cornerstone of HIV care, decreases the incidence of TB in HIV-uninfected people, this remains 4- to 7-fold higher after ART in HIV-co-infected individuals in TB-endemic settings, regardless of the duration of therapy. Thus, the immune control of Mtb infection in Mtb/HIV-co-infected individuals is not fully restored by ART. We do not fully understand the reasons why Mtb/HIV-co-infected individuals maintain a high susceptibility to the reactivation of LTBI, despite an effective viral control by ART. A deep understanding of the molecular mechanisms that govern HIV-induced reactivation of TB is essential to develop improved treatments and vaccines for the Mtb/HIV-co-infected population. We discuss potential strategies for the mitigation of the observed chronic immune activation in combination with both anti-TB and anti-retroviral approaches.
Collapse
Affiliation(s)
| | | | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| |
Collapse
|
11
|
Azevedo-Pereira JM, Pires D, Calado M, Mandal M, Santos-Costa Q, Anes E. HIV/Mtb Co-Infection: From the Amplification of Disease Pathogenesis to an “Emerging Syndemic”. Microorganisms 2023; 11:microorganisms11040853. [PMID: 37110276 PMCID: PMC10142195 DOI: 10.3390/microorganisms11040853] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Human immunodeficiency virus (HIV) and Mycobacterium tuberculosis (Mtb) are pathogens responsible for millions of new infections each year; together, they cause high morbidity and mortality worldwide. In addition, late-stage HIV infection increases the risk of developing tuberculosis (TB) by a factor of 20 in latently infected people, and even patients with controlled HIV infection on antiretroviral therapy (ART) have a fourfold increased risk of developing TB. Conversely, Mtb infection exacerbates HIV pathogenesis and increases the rate of AIDS progression. In this review, we discuss this reciprocal amplification of HIV/Mtb coinfection and how they influence each other’s pathogenesis. Elucidating the infectious cofactors that impact on pathogenesis may open doors for the design of new potential therapeutic strategies to control disease progression, especially in contexts where vaccines or the sterile clearance of pathogens are not effectively available.
Collapse
Affiliation(s)
- José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence: (J.M.A.-P.); (E.A.)
| | - David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Estrada Octávio Pato, 2635-631 Rio de Mouro, Portugal
| | - Marta Calado
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Manoj Mandal
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Quirina Santos-Costa
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence: (J.M.A.-P.); (E.A.)
| |
Collapse
|
12
|
Singh B, Moodley C, Singh DK, Escobedo RA, Sharan R, Arora G, Ganatra SR, Shivanna V, Gonzalez O, Hall-Ursone S, Dick EJ, Kaushal D, Alvarez X, Mehra S. Inhibition of indoleamine dioxygenase leads to better control of tuberculosis adjunctive to chemotherapy. JCI Insight 2023; 8:e163101. [PMID: 36692017 PMCID: PMC9977315 DOI: 10.1172/jci.insight.163101] [Citation(s) in RCA: 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: 07/05/2022] [Accepted: 11/30/2022] [Indexed: 01/24/2023] Open
Abstract
The expression of indoleamine 2,3-dioxygenase (IDO), a robust immunosuppressant, is significantly induced in macaque tuberculosis (TB) granulomas, where it is expressed on IFN-responsive macrophages and myeloid-derived suppressor cells. IDO expression is also highly induced in human TB granulomas, and products of its activity are detected in patients with TB. In vivo blockade of IDO activity resulted in the reorganization of the granuloma with substantially greater T cells being recruited to the core of the lesions. This correlated with better immune control of TB and reduced lung M. tuberculosis burdens. To study if the IDO blockade strategy can be translated to a bona fide host-directed therapy in the clinical setting of TB, we studied the effect of IDO inhibitor 1-methyl-d-tryptophan adjunctive to suboptimal anti-TB chemotherapy. While two-thirds of controls and one-third of chemotherapy-treated animals progressed to active TB, inhibition of IDO adjunctive to the same therapy protected macaques from TB, as measured by clinical, radiological, and microbiological attributes. Although chemotherapy improved proliferative T cell responses, adjunctive inhibition of IDO further enhanced the recruitment of effector T cells to the lung. These results strongly suggest the possibility that IDO inhibition can be attempted adjunctive to anti-TB chemotherapy in clinical trials.
Collapse
|
13
|
Walker EM, Merino KM, Slisarenko N, Grasperge BF, Mehra S, Roy CJ, Kaushal D, Rout N. Impact of SIV infection on mycobacterial lipid-reactive T cell responses in Bacillus Calmette-Guérin (BCG) inoculated macaques. Front Immunol 2023; 13:1085786. [PMID: 36726992 PMCID: PMC9885173 DOI: 10.3389/fimmu.2022.1085786] [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: 10/31/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Background Although BCG vaccine protects infants from tuberculosis (TB), it has limited efficacy in adults against pulmonary TB. Further, HIV coinfection significantly increases the risk of developing active TB. In the lack of defined correlates of protection in TB disease, it is essential to explore immune responses beyond conventional CD4 T cells to gain a better understanding of the mechanisms of TB immunity. Methods Here, we evaluated unconventional lipid-reactive T cell responses in cynomolgus macaques following aerosol BCG inoculation and examined the impact of subsequent SIV infection on these responses. Immune responses to cellular lipids of M. bovis and M. tuberculosis were examined ex vivo in peripheral blood and bronchioalveolar lavage (BAL). Results Prior to BCG inoculation, innate-like IFN-γ responses to mycobacterial lipids were observed in T cells. Aerosol BCG exposure induced an early increase in frequencies of BAL γδT cells, a dominant subset of lipid-reactive T cells, along with enhanced IL-7R and CXCR3 expression. Further, BCG exposure stimulated greater IFN-γ responses to mycobacterial lipids in peripheral blood and BAL, suggesting the induction of systemic and local Th1-type response in lipid-reactive T cells. Subsequent SIV infection resulted in a significant loss of IL-7R expression on blood and BAL γδT cells. Additionally, IFN-γ responses of mycobacterial lipid-reactive T cells in BAL fluid were significantly lower in SIV-infected macaques, while perforin production was maintained through chronic SIV infection. Conclusions Overall, these data suggest that despite SIV-induced decline in IL-7R expression and IFN-γ production by mycobacterial lipid-reactive T cells, their cytolytic potential is maintained. A deeper understanding of anti-mycobacterial lipid-reactive T cell functions may inform novel approaches to enhance TB control in individuals with or without HIV infection.
Collapse
Affiliation(s)
- Edith M. Walker
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
| | - Kristen M. Merino
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
| | - Nadia Slisarenko
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
| | - Brooke F. Grasperge
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Chad J. Roy
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Namita Rout
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
14
|
Immune cell interactions in tuberculosis. Cell 2022; 185:4682-4702. [PMID: 36493751 DOI: 10.1016/j.cell.2022.10.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 12/13/2022]
Abstract
Despite having been identified as the organism that causes tuberculosis in 1882, Mycobacterium tuberculosis has managed to still evade our understanding of the protective immune response against it, defying the development of an effective vaccine. Technology and novel experimental models have revealed much new knowledge, particularly with respect to the heterogeneity of the bacillus and the host response. This review focuses on certain immunological elements that have recently yielded exciting data and highlights the importance of taking a holistic approach to understanding the interaction of M. tuberculosis with the many host cells that contribute to the development of protective immunity.
Collapse
|
15
|
Hoerter A, Arnett E, Schlesinger LS, Pienaar E. Systems biology approaches to investigate the role of granulomas in TB-HIV coinfection. Front Immunol 2022; 13:1014515. [PMID: 36405707 PMCID: PMC9670175 DOI: 10.3389/fimmu.2022.1014515] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/20/2022] [Indexed: 09/29/2023] Open
Abstract
The risk of active tuberculosis disease is 15-21 times higher in those coinfected with human immunodeficiency virus-1 (HIV) compared to tuberculosis alone, and tuberculosis is the leading cause of death in HIV+ individuals. Mechanisms driving synergy between Mycobacterium tuberculosis (Mtb) and HIV during coinfection include: disruption of cytokine balances, impairment of innate and adaptive immune cell functionality, and Mtb-induced increase in HIV viral loads. Tuberculosis granulomas are the interface of host-pathogen interactions. Thus, granuloma-based research elucidating the role and relative impact of coinfection mechanisms within Mtb granulomas could inform cohesive treatments that target both pathogens simultaneously. We review known interactions between Mtb and HIV, and discuss how the structure, function and development of the granuloma microenvironment create a positive feedback loop favoring pathogen expansion and interaction. We also identify key outstanding questions and highlight how coupling computational modeling with in vitro and in vivo efforts could accelerate Mtb-HIV coinfection discoveries.
Collapse
Affiliation(s)
- Alexis Hoerter
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Eusondia Arnett
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Larry S. Schlesinger
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
16
|
Gough M, Singh DK, Singh B, Kaushal D, Mehra S. System-wide identification of myeloid markers of TB disease and HIV-induced reactivation in the macaque model of Mtb infection and Mtb/SIV co-infection. Front Immunol 2022; 13:777733. [PMID: 36275677 PMCID: PMC9583676 DOI: 10.3389/fimmu.2022.777733] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) has developed specialized mechanisms to parasitize its host cell, the macrophage. These mechanisms allow it to overcome killing by oxidative burst and persist in the wake of an inflammatory response. Mtb infection in the majority of those exposed is controlled in an asymptomatic form referred to as latent tuberculosis infection (LTBI). HIV is a well-known catalyst of reactivation of LTBI to active TB infection (ATB). Through the use of nonhuman primates (NHPs) co-infected with Mtb and Simian Immunodeficiency Virus (Mtb/SIV), we are able to simulate human progression of TB/AIDS comorbidity. The advantage of NHP models is that they recapitulate the breadth of human TB outcomes, including immune control of infection, and loss of this control due to SIV co-infection. Identifying correlates of immune control of infection is important for both vaccine and therapeutics development. Using macaques infected with Mtb or Mtb/SIV and with different clinical outcomes we attempted to identify signatures between those that progress to active infection after SIV challenge (reactivators) and those that control the infection (non-reactivators). We particularly focused on pathways relevant to myeloid origin cells such as macrophages, as these innate immunocytes have an important contribution to the initial control or the lack thereof, following Mtb infection. Using bacterial burden, C-reactive protein (CRP), and other clinical indicators of disease severity as a guide, we were able to establish gene signatures of host disease state and progression. In addition to gene signatures, clustering algorithms were used to differentiate between host disease states and identify relationships between genes. This allowed us to identify clusters of genes which exhibited differential expression profiles between the three groups of macaques: ATB, LTBI and Mtb/SIV. The gene signatures were associated with pathways relevant to apoptosis, ATP production, phagocytosis, cell migration, and Type I interferon (IFN), which are related to macrophage function. Our results suggest novel macrophage functions that may play roles in the control of Mtb infection with and without co-infection with SIV. These results particularly point towards an interplay between Type I IFN signaling and IFN-γ signaling, and the resulting impact on lung macrophages as an important determinant of progression to TB.
Collapse
|
17
|
Sharan R, Ganatra SR, Singh DK, Cole J, Foreman TW, Thippeshappa R, Peloquin CA, Shivanna V, Gonzalez O, Day CL, Gandhi NR, Dick EJ, Hall-Ursone S, Mehra S, Schlesinger LS, Rengarajan J, Kaushal D. Isoniazid and rifapentine treatment effectively reduces persistent M. tuberculosis infection in macaque lungs. J Clin Invest 2022; 132:e161564. [PMID: 35862216 PMCID: PMC9479578 DOI: 10.1172/jci161564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/19/2022] [Indexed: 02/01/2023] Open
Abstract
A once-weekly oral dose of isoniazid and rifapentine for 3 months (3HP) is recommended by the CDC for treatment of latent tuberculosis infection (LTBI). The aim of this study is to assess 3HP-mediated clearance of M. tuberculosis bacteria in macaques with asymptomatic LTBI. Twelve Indian-origin rhesus macaques were infected with a low dose (~10 CFU) of M. tuberculosis CDC1551 via aerosol. Six animals were treated with 3HP and 6 were left untreated. The animals were imaged via PET/CT at frequent intervals. Upon treatment completion, all animals except 1 were coinfected with SIV to assess reactivation of LTBI to active tuberculosis (ATB). Four of 6 treated macaques showed no evidence of persistent bacilli or extrapulmonary spread until the study end point. PET/CT demonstrated the presence of significantly more granulomas in untreated animals relative to the treated group. The untreated animals harbored persistent bacilli and demonstrated tuberculosis (TB) reactivation following SIV coinfection, while none of the treated animals reactivated to ATB. 3HP treatment effectively reduced persistent infection with M. tuberculosis and prevented reactivation of TB in latently infected macaques.
Collapse
Affiliation(s)
- Riti Sharan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Shashank R. Ganatra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Dhiraj K. Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Journey Cole
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Taylor W. Foreman
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Rajesh Thippeshappa
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | | | - Vinay Shivanna
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Olga Gonzalez
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | | | - Neel R. Gandhi
- Emory Tuberculosis Center and
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Edward J. Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Shannan Hall-Ursone
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Larry S. Schlesinger
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jyothi Rengarajan
- Emory Tuberculosis Center and
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
18
|
Scioscia G, Lacedonia D, Giuffreda E, Caccavo I, Quarato CMI, Soccio P, Tondo P, Sassani EV, Pescatore D, Foschino Barbaro MP. Adaptive immunity in different CT patterns of active tuberculosis and possible variability according to patients' geographic provenience. Front Med (Lausanne) 2022; 9:890609. [PMID: 36160177 PMCID: PMC9489992 DOI: 10.3389/fmed.2022.890609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIt is still unclear if low lymphocyte levels are directly related to immunological modifications induced by the TB infection or if they depend on the general pre-existing health impairment of affected patients. Our aim was to detect eventual differences in the immunological status of patients with pulmonary TB compared to an age and sex-matched group of hospitalized patients with other bacterial community-acquired pneumonia (CAP). In addition, we tried to assess an association between alterations in the peripheral lymphocyte subsets and the development of different CT patterns of active TB and to discover differences in the immunological status and in the radiological patterns of TB presentation between patients of different geographic proveniences.MethodsThis observational study included 48 patients with TB and 48 sex- and age-matched patients affected by other bacterial CAP. The presence of HIV/AIDS, other immunocompromising conditions, and confounding chronic pulmonary comorbidities was excluded. Flow cytometry was performed on all the enrolled subjects at admission, before starting the appropriate antibiotic therapy. Patients with TB also underwent a computed tomography (CT) scan.ResultsPatients with TB showed a decrease in the absolute count of all the lymphocyte subsets compared to the CAP group. Only the reduction in the percentage of CD4+ T-lymphocytes was significant, while the percentage of CD8+ T-lymphocytes was significantly increased. Patients presenting exudative forms with atypical locations of TB showed a significant reduction in the absolute count and percentage of CD19+ B-lymphocytes compared to those affected by productive TB forms with the typical location. Despite being younger, our black Sub-Saharan Africans showed a significant reduction in the CD4+ T-lymphocytes compartment and a higher prevalence of atypical and exudative forms of TB compared with white Europeans.ConclusionTuberculosis itself may alter peripheral blood lymphocyte subsets compared to other CAP. An impaired CD19+ B-lymphocyte compartment may result in an abnormal exudative response in atypical locations and a suboptimal bacterial control. Other constitutive or environmental causes may influence immunological differences found in patients with TB, particularly in case of different geographic origins. Anyhow, flow cytometry may be of great value in evaluating the immune function of these patients.
Collapse
Affiliation(s)
- Giulia Scioscia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, Foggia, Italy
| | - Donato Lacedonia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, Foggia, Italy
| | - Ernesto Giuffreda
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, Foggia, Italy
| | - Incoronata Caccavo
- Internistic Department, Institute of Respiratory Disease, Azienda Ospedaliera Regionale San Carlo, Potenza, Italy
| | - Carla Maria Irene Quarato
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, Foggia, Italy
- *Correspondence: Carla Maria Irene Quarato
| | - Piera Soccio
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Pasquale Tondo
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | | | - Dalila Pescatore
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Maria Pia Foschino Barbaro
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, Foggia, Italy
| |
Collapse
|
19
|
Bucşan AN, Veatch A, Singh DK, Akter S, Golden NA, Kirkpatrick M, Threeton B, Moodley C, Ahmed M, Doyle LA, Russell-Lodrigue K, Norton EB, Didier PJ, Roy CJ, Abramovitch RB, Mehra S, Khader SA, Kaushal D. Response to Hypoxia and the Ensuing Dysregulation of Inflammation Impacts Mycobacterium tuberculosis Pathogenicity. Am J Respir Crit Care Med 2022; 206:94-104. [PMID: 35412961 PMCID: PMC9718519 DOI: 10.1164/rccm.202112-2747oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Different Mycobacterium tuberculosis (Mtb) strains exhibit variable degrees of virulence in humans and animal models. Differing stress response strategies used by different strains of Mtb could influence virulence. Objectives: We compared the virulence of two strains of Mtb with use in animal model research: CDC1551 and Erdman. Methods: Rhesus macaques, which develop human-like tuberculosis attributes and pathology, were infected with a high dose of either strain via aerosol, and virulence was compared by bacterial burden and pathology. Measurements and Main Results: Infection with Erdman resulted in significantly shorter times to euthanasia and higher bacterial burdens and greater systemic inflammation and lung pathology relative to those infected with CDC1551. Macaques infected with Erdman also exhibited significantly higher early inflammatory myeloid cell influx to the lung, greater macrophage and T cell activity, and higher expression of lung remodeling (extracellular matrix) genes, consistent with greater pathology. Expression of NOTCH4 (neurogenic locus notch homolog 4) signaling, which is induced in response to hypoxia and promotes undifferentiated cellular state, was also higher in Erdman-infected lungs. The granulomas generated by Erdman, and not CDC1551, infection appeared to have larger regions of necrosis, which is strongly associated with hypoxia. To better understand the mechanisms of differential hypoxia induction by these strains, we subjected both to hypoxia in vitro. Erdman induced higher concentrations of DosR regulon relative to CDC1551. The DosR regulon is the global regulator of response to hypoxia in Mtb and critical for its persistence in granulomas. Conclusions: Our results show that the response to hypoxia is a critical mediator of virulence determination in Mtb, with potential impacts on bacillary persistence, reactivation, and efficiency of therapeutics.
Collapse
Affiliation(s)
- Allison N. Bucşan
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Ashley Veatch
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Dhiraj K. Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Sadia Akter
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Nadia A. Golden
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Melanie Kirkpatrick
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Breanna Threeton
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Chivonne Moodley
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Mushtaq Ahmed
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Lara A. Doyle
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Kasi Russell-Lodrigue
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Elizabeth B. Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana; and
| | - Peter J. Didier
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
| | - Chad J. Roy
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Robert B. Abramovitch
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan
| | - Smriti Mehra
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Deepak Kaushal
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| |
Collapse
|
20
|
Moriarty RV, Rodgers MA, Ellis AL, Balgeman AJ, Larson EC, Hopkins F, Chase MR, Maiello P, Fortune SM, Scanga CA, O’Connor SL. Spontaneous Control of SIV Replication Does Not Prevent T Cell Dysregulation and Bacterial Dissemination in Animals Co-Infected with M. tuberculosis. Microbiol Spectr 2022; 10:e0172421. [PMID: 35467372 PMCID: PMC9241861 DOI: 10.1128/spectrum.01724-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 04/08/2022] [Indexed: 12/27/2022] Open
Abstract
Individuals co-infected with HIV and Mycobacterium tuberculosis (Mtb) are more likely to develop severe tuberculosis (TB) disease than HIV-naive individuals. To understand how a chronic pre-existing Simian immunodeficiency virus (SIV) infection impairs the early immune response to Mtb, we used the Mauritian cynomolgus macaque (MCM) model of SIV/Mtb co-infection. We examined the relationship between peripheral viral control and Mtb burden, Mtb dissemination, and T cell function between SIV+ spontaneous controllers, SIV+ non-controllers, and SIV-naive MCM who were challenged with a barcoded Mtb Erdman strain 6 months post-SIV infection and necropsied 6 weeks post-Mtb infection. Mycobacterial burden was highest in the SIV+ non-controllers in all assessed tissues. In lung granulomas, the frequency of TNF-α-producing CD4+ T cells was reduced in all SIV+ MCM, but IFNγ-producing CD4+ T cells were only lower in the SIV+ non-controllers. Further, while all SIV+ MCM had more PD1+ and TIGIT+ T cells in the lung granulomas relative to SIV-naive MCM, SIV+ controllers exhibited the highest frequency of cells expressing these markers. To measure the effect of SIV infection on within-host bacterial dissemination, we sequenced the molecular barcodes of Mtb present in each tissue and characterized the Mtb population complexity. While Mtb population complexity was not associated with SIV infection group, lymph nodes had increased complexity when compared with lung granulomas across all groups. These results provide evidence that SIV+ animals, independent of viral control, exhibit a dysregulated T cell immune response and enhanced dissemination of Mtb, likely contributing to the poor TB disease course across all SIV/Mtb co-infected animals. IMPORTANCE HIV and TB remain significant global health issues, despite the availability of treatments. Individuals with HIV, including those who are virally suppressed, are at an increased risk to develop and succumb to severe TB disease when compared with HIV-naive individuals. Our study aims to understand the relationship between the extent of SIV replication, mycobacterial growth, and T cell function in the tissues of co-infected Mauritian cynomolgus macaques during the first 6 weeks of Mtb infection. Here we demonstrate that increased viral replication is associated with increased bacterial burden in the tissues and impaired T cell responses, and that the immunological damage attributed to virus infection is not fully eliminated when animals spontaneously control virus replication.
Collapse
Affiliation(s)
- Ryan V. Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mark A. Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy L. Ellis
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alexis J. Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Erica C. Larson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Forrest Hopkins
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Michael R. Chase
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
21
|
Shu Q, Liu S, Alonzi T, LaCourse SM, Singh DK, Bao D, Wamalwa D, Jiang L, Lyon CJ, John-Stewart G, Kaushal D, Goletti D, Hu T. Assay design for unambiguous identification and quantification of circulating pathogen-derived peptide biomarkers. Theranostics 2022; 12:2948-2962. [PMID: 35401822 PMCID: PMC8965485 DOI: 10.7150/thno.70373] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/25/2022] [Indexed: 11/05/2022] Open
Abstract
Rationale: Circulating pathogen-derived proteins can serve as useful biomarkers for infections but may be detected with poor sensitivity and specificity by standard immunoassays due to masking effects and cross-reactivity. Mass spectrometry (MS)-read immunoassays for biomarker-derived peptides can resolve these issues, but lack standard workflows to select species-specific peptides with strong MS signal that are suitable for antibody generation. Methods:Using a Mycobacterium tuberculosis (Mtb) protein as an example, candidate peptides were selected by length, species-specificity, MS intensity, and antigenicity score. MS data from spiked healthy serum was employed to define MS feature thresholds, including a novel measure of internal MS data correlation, to produce a peak detection algorithm. Results: This algorithm performed better in rejecting false positive signal than each of its criteria, including those currently employed for this purpose. Analysis of an Mtb peptide biomarker (CFP-10pep) by this approach identified tuberculosis cases not detected by microbiologic assays, including extrapulmonary tuberculosis and tuberculosis cases in children infected with HIV-1. Circulating CFP-10pep levels measured in a non-human primate model of tuberculosis distinguished disease from asymptomatic infection and tended to correspond with Mtb granuloma size, suggesting that it could also serve as a surrogate marker for Mtb burden and possibly treatment response. Conclusions: These biomarker selection and analysis approach appears to have strong potential utility for infectious disease diagnosis, including cryptic infections, and possibly to monitor changes in Mtb burden that may reflect disease progression or a response to treatment, which are critical needs for more effective disease control.
Collapse
Affiliation(s)
- Qingbo Shu
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Shan Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Medical Genetics, Department of Laboratory medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Tonino Alonzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Sylvia M. LaCourse
- Departments of Medicine, Division of Allergy and Infectious Diseases, and Global Health, University of Washington, Seattle, USA
| | - Dhiraj Kumar Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Duran Bao
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Dalton Wamalwa
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Li Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Medical Genetics, Department of Laboratory medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Christopher J. Lyon
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Grace John-Stewart
- Departments of Medicine, Division of Allergy and Infectious Diseases, and Global Health, University of Washington, Seattle, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Tony Hu
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, Louisiana, USA.,✉ Corresponding author: Tony Hu.
| |
Collapse
|
22
|
SIV Evolutionary Dynamics in Cynomolgus Macaques during SIV- Mycobacterium tuberculosis Co-Infection. Viruses 2021; 14:v14010048. [PMID: 35062252 PMCID: PMC8778162 DOI: 10.3390/v14010048] [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: 10/15/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 01/08/2023] Open
Abstract
Co-infection with Mycobacterium tuberculosis (Mtb) and human immunodeficiency virus (HIV) is a worldwide public health concern, leading to worse clinical outcomes caused by both pathogens. We used a non-human primate model of simian immunodeficiency virus (SIV)-Mtb co-infection, in which latent Mtb infection was established prior to SIVmac251 infection. The evolutionary dynamics of SIV env was evaluated from samples in plasma, lymph nodes, and lungs (including granulomas) of SIV-Mtb co-infected and SIV only control animals. While the diversity of the challenge virus was low and overall viral diversity remained relatively low over 6–9 weeks, changes in viral diversity and divergence were observed, including evidence for tissue compartmentalization. Overall, viral diversity was highest in SIV-Mtb animals that did not develop clinical Mtb reactivation compared to animals with Mtb reactivation. Among lung granulomas, viral diversity was positively correlated with the frequency of CD4+ T cells and negatively correlated with the frequency of CD8+ T cells. SIV diversity was highest in the thoracic lymph nodes compared to other sites, suggesting that lymphatic drainage from the lungs in co-infected animals provides an advantageous environment for SIV replication. This is the first assessment of SIV diversity across tissue compartments during SIV-Mtb co-infection after established Mtb latency.
Collapse
|
23
|
Sharan R, Ganatra SR, Bucsan AN, Cole J, Singh DK, Alvarez X, Gough M, Alvarez C, Blakley A, Ferdin J, Thippeshappa R, Singh B, Escobedo R, Shivanna V, Dick EJ, Hall-Ursone S, Khader SA, Mehra S, Rengarajan J, Kaushal D. Antiretroviral therapy timing impacts latent tuberculosis infection reactivation in a tuberculosis/simian immunodeficiency virus coinfection model. J Clin Invest 2021; 132:153090. [PMID: 34855621 PMCID: PMC8803324 DOI: 10.1172/jci153090] [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: 07/12/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022] Open
Abstract
Studies using the nonhuman primate model of Mycobacteriumtuberculosis/simian immunodeficiency virus coinfection have revealed protective CD4+ T cell–independent immune responses that suppress latent tuberculosis infection (LTBI) reactivation. In particular, chronic immune activation rather than the mere depletion of CD4+ T cells correlates with reactivation due to SIV coinfection. Here, we administered combinatorial antiretroviral therapy (cART) 2 weeks after SIV coinfection to study whether restoration of CD4+ T cell immunity occurred more broadly, and whether this prevented reactivation of LTBI compared to cART initiated 4 weeks after SIV. Earlier initiation of cART enhanced survival, led to better control of viral replication, and reduced immune activation in the periphery and lung vasculature, thereby reducing the rate of SIV-induced reactivation. We observed robust CD8+ T effector memory responses and significantly reduced macrophage turnover in the lung tissue. However, skewed CD4+ T effector memory responses persisted and new TB lesions formed after SIV coinfection. Thus, reactivation of LTBI is governed by very early events of SIV infection. Timing of cART is critical in mitigating chronic immune activation. The potential novelty of these findings mainly relates to the development of a robust animal model of human M. tuberculosis/HIV coinfection that allows the testing of underlying mechanisms.
Collapse
Affiliation(s)
- Riti Sharan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Shashank R Ganatra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Allison N Bucsan
- Department of Molecular Microbiology, Washington University, St. Louis, St. Louis, United States of America
| | - Journey Cole
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Dhiraj K Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Xavier Alvarez
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Maya Gough
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Cynthia Alvarez
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Alyssa Blakley
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Justin Ferdin
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Rajesh Thippeshappa
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Bindu Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Ruby Escobedo
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Vinay Shivanna
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Edward J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Shannan Hall-Ursone
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University, St. Louis, St. Louis, United States of America
| | - Smriti Mehra
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, United States of America
| | - Jyothi Rengarajan
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, United States of America
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, United States of America
| |
Collapse
|
24
|
Boom WH, Schaible UE, Achkar JM. The knowns and unknowns of latent Mycobacterium tuberculosis infection. J Clin Invest 2021; 131:136222. [PMID: 33529162 DOI: 10.1172/jci136222] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Humans have been infected with Mycobacterium tuberculosis (Mtb) for thousands of years. While tuberculosis (TB), one of the deadliest infectious diseases, is caused by uncontrolled Mtb infection, over 90% of presumed infected individuals remain asymptomatic and contain Mtb in a latent TB infection (LTBI) without ever developing disease, and some may clear the infection. A small number of heavily Mtb-exposed individuals appear to resist developing traditional LTBI. Because Mtb has mechanisms for intracellular survival and immune evasion, successful control involves all of the arms of the immune system. Here, we focus on immune responses to Mtb in humans and nonhuman primates and discuss new concepts and outline major knowledge gaps in our understanding of LTBI, ranging from the earliest events of exposure and infection to success or failure of Mtb control.
Collapse
Affiliation(s)
- W Henry Boom
- Department of Medicine.,Department of Pathology, and.,Department of Molecular Biology and Microbiology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Ulrich E Schaible
- Division of Cellular Microbiology, Research Center Borstel-Leibniz Lung Center, Borstel, Germany.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Jacqueline M Achkar
- Department of Medicine and.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
25
|
Lemaitre J, Naninck T, Delache B, Creppy J, Huber P, Holzapfel M, Bouillier C, Contreras V, Martinon F, Kahlaoui N, Pascal Q, Tricot S, Ducancel F, Vecellio L, Le Grand R, Maisonnasse P. Non-human primate models of human respiratory infections. Mol Immunol 2021; 135:147-164. [PMID: 33895579 PMCID: PMC8062575 DOI: 10.1016/j.molimm.2021.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/03/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022]
Abstract
Respiratory pathogens represent a great burden for humanity and a potential source of new pandemics, as illustrated by the recent emergence of coronavirus disease 2019 (COVID-19). In recent decades, biotechnological advances have led to the development of numerous innovative therapeutic molecules and vaccine immunogens. However, we still lack effective treatments and vaccines against many respiratory pathogens. More than ever, there is a need for a fast, predictive, preclinical pipeline, to keep pace with emerging diseases. Animal models are key for the preclinical development of disease management strategies. The predictive value of these models depends on their ability to reproduce the features of the human disease, the mode of transmission of the infectious agent and the availability of technologies for monitoring infection. This review focuses on the use of non-human primates as relevant preclinical models for the development of prevention and treatment for human respiratory infections.
Collapse
Affiliation(s)
- Julien Lemaitre
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Thibaut Naninck
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Benoît Delache
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Justina Creppy
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France; Centre d'Etude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Philippe Huber
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Marion Holzapfel
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Camille Bouillier
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Vanessa Contreras
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Frédéric Martinon
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Nidhal Kahlaoui
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Quentin Pascal
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Sabine Tricot
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Frédéric Ducancel
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Laurent Vecellio
- Centre d'Etude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France; Plateforme Scientifique et Technique Animaleries (PST-A), Université de Tours, Tours, France
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Pauline Maisonnasse
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France.
| |
Collapse
|
26
|
Larson EC, Ellis-Connell A, Rodgers MA, Balgeman AJ, Moriarty RV, Ameel CL, Baranowski TM, Tomko JA, Causgrove CM, Maiello P, O'Connor SL, Scanga CA. Pre-existing Simian Immunodeficiency Virus Infection Increases Expression of T Cell Markers Associated with Activation during Early Mycobacterium tuberculosis Coinfection and Impairs TNF Responses in Granulomas. THE JOURNAL OF IMMUNOLOGY 2021; 207:175-188. [PMID: 34145063 DOI: 10.4049/jimmunol.2100073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/19/2021] [Indexed: 01/01/2023]
Abstract
Tuberculosis (TB) is the leading infectious cause of death among people living with HIV. People living with HIV are more susceptible to contracting Mycobacterium tuberculosis and often have worsened TB disease. Understanding the immunologic defects caused by HIV and the consequences it has on M. tuberculosis coinfection is critical in combating this global health epidemic. We previously showed in a model of SIV and M. tuberculosis coinfection in Mauritian cynomolgus macaques (MCM) that SIV/M. tuberculosis-coinfected MCM had rapidly progressive TB. We hypothesized that pre-existing SIV infection impairs early T cell responses to M. tuberculosis infection. We infected MCM with SIVmac239, followed by coinfection with M. tuberculosis Erdman 6 mo later. Although similar, TB progression was observed in both SIV+ and SIV-naive animals at 6 wk post-M. tuberculosis infection; longitudinal sampling of the blood (PBMC) and airways (bronchoalveolar lavage) revealed a significant reduction in circulating CD4+ T cells and an influx of CD8+ T cells in airways of SIV+ animals. At sites of M. tuberculosis infection (i.e., granulomas), SIV/M. tuberculosis-coinfected animals had a higher proportion of CD4+ and CD8+ T cells expressing PD-1 and TIGIT. In addition, there were fewer TNF-producing CD4+ T cells in granulomas of SIV/M. tuberculosis-coinfected animals. Taken together, we show that concurrent SIV infection alters T cell phenotypes in granulomas during the early stages of TB disease. As it is critical to establish control of M. tuberculosis replication soon postinfection, these phenotypic changes may distinguish the immune dysfunction that arises from pre-existing SIV infection, which promotes TB progression.
Collapse
Affiliation(s)
- Erica C Larson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA;
| | - Amy Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, WI
| | - Mark A Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Alexis J Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, WI
| | - Ryan V Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, WI
| | - Cassaundra L Ameel
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Tonilynn M Baranowski
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jaime A Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Chelsea M Causgrove
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, WI.,Wisconsin National Primate Research Center, University of Wisconsin-Madison, WI; and
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA; .,Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
27
|
Moriarty RV, Ellis AL, O’Connor SL. Monkeying around with MAIT Cells: Studying the Role of MAIT Cells in SIV and Mtb Co-Infection. Viruses 2021; 13:863. [PMID: 34066765 PMCID: PMC8151491 DOI: 10.3390/v13050863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 11/25/2022] Open
Abstract
There were an estimated 10 million new cases of tuberculosis (TB) disease in 2019. While over 90% of individuals successfully control Mycobacterium tuberculosis (Mtb) infection, which causes TB disease, HIV co-infection often leads to active TB disease. Despite the co-endemic nature of HIV and TB, knowledge of the immune mechanisms contributing to the loss of control of Mtb replication during HIV infection is lacking. Mucosal-associated invariant T (MAIT) cells are innate-like T cells that target and destroy bacterially-infected cells and may contribute to the control of Mtb infection. Studies examining MAIT cells in human Mtb infection are commonly performed using peripheral blood samples. However, because Mtb infection occurs primarily in lung tissue and lung-associated lymph nodes, these studies may not be fully translatable to the tissues. Additionally, studies longitudinally examining MAIT cell dynamics during HIV/Mtb co-infection are rare, and lung and lymph node tissue samples from HIV+ patients are typically unavailable. Nonhuman primates (NHP) provide a model system to characterize MAIT cell activity during Mtb infection, both in Simian Immunodeficiency Virus (SIV)-infected and SIV-naïve animals. Using NHPs allows for a more comprehensive understanding of tissue-based MAIT cell dynamics during infection with both pathogens. NHP SIV and Mtb infection is similar to human HIV and Mtb infection, and MAIT cells are phenotypically similar in humans and NHPs. Here, we discuss current knowledge surrounding MAIT cells in SIV and Mtb infection, how SIV infection impairs MAIT cell function during Mtb co-infection, and knowledge gaps to address.
Collapse
Affiliation(s)
| | | | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA; (R.V.M.); (A.L.E.)
| |
Collapse
|
28
|
Cronan MR, Hughes EJ, Brewer WJ, Viswanathan G, Hunt EG, Singh B, Mehra S, Oehlers SH, Gregory SG, Kaushal D, Tobin DM. A non-canonical type 2 immune response coordinates tuberculous granuloma formation and epithelialization. Cell 2021; 184:1757-1774.e14. [PMID: 33761328 PMCID: PMC8055144 DOI: 10.1016/j.cell.2021.02.046] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 11/03/2020] [Accepted: 02/22/2021] [Indexed: 12/19/2022]
Abstract
The central pathogen-immune interface in tuberculosis is the granuloma, a complex host immune structure that dictates infection trajectory and physiology. Granuloma macrophages undergo a dramatic transition in which entire epithelial modules are induced and define granuloma architecture. In tuberculosis, relatively little is known about the host signals that trigger this transition. Using the zebrafish-Mycobacterium marinum model, we identify the basis of granuloma macrophage transformation. Single-cell RNA-sequencing analysis of zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected macaques reveal that, even in the presence of robust type 1 immune responses, countervailing type 2 signals associate with macrophage epithelialization. We find that type 2 immune signaling, mediated via stat6, is absolutely required for epithelialization and granuloma formation. In mixed chimeras, stat6 acts cell autonomously within macrophages, where it is required for epithelioid transformation and incorporation into necrotic granulomas. These findings establish the signaling pathway that produces the hallmark structure of mycobacterial infection.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified/genetics
- Animals, Genetically Modified/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation
- Disease Models, Animal
- Epithelioid Cells/cytology
- Epithelioid Cells/immunology
- Epithelioid Cells/metabolism
- Granuloma/immunology
- Granuloma/metabolism
- Granuloma/pathology
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/metabolism
- Immunity/physiology
- Interferon-gamma/metabolism
- Interleukin-12/metabolism
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Mycobacterium Infections, Nontuberculous/immunology
- Mycobacterium Infections, Nontuberculous/pathology
- Mycobacterium marinum/isolation & purification
- Mycobacterium marinum/physiology
- Necrosis
- Receptors, Interleukin-4/antagonists & inhibitors
- Receptors, Interleukin-4/genetics
- Receptors, Interleukin-4/metabolism
- STAT6 Transcription Factor/antagonists & inhibitors
- STAT6 Transcription Factor/genetics
- STAT6 Transcription Factor/metabolism
- Signal Transduction
- Zebrafish/growth & development
- Zebrafish/metabolism
- RNA, Guide, CRISPR-Cas Systems
Collapse
Affiliation(s)
- Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Erika J Hughes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; University Program in Genetics and Genomics, Duke University School of Medicine, Durham, NC 27710, USA
| | - W Jared Brewer
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gopinath Viswanathan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Emily G Hunt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bindu Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia; The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
29
|
Pillay K, Lewis L, Rambaran S, Yende-Zuma N, Archary D, Gengiah S, Govender D, Hassan-Moosa R, Samsunder N, Abdool Karim SS, McKinnon LR, Padayatchi N, Naidoo K, Sivro A. Plasma Biomarkers of Risk of Tuberculosis Recurrence in HIV Co-Infected Patients From South Africa. Front Immunol 2021; 12:631094. [PMID: 33841412 PMCID: PMC8026888 DOI: 10.3389/fimmu.2021.631094] [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: 11/19/2020] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
There is an urgent need to identify immunological markers of tuberculosis (TB) risk in HIV co-infected individuals. Previously we have shown that TB recurrence in HIV co-infected individuals on ART was associated with markers of systemic inflammation (IL-6, IL1β and IL-1Rα). Here we examined the effect of additional acute inflammation and microbial translocation marker expression on risk of TB recurrence. Stored plasma samples were drawn from the TB Recurrence upon Treatment with HAART (TRuTH) study, in which individuals with previously treated pulmonary TB were screened for recurrence quarterly for up to 4 years. Recurrent TB cases (n = 37) were matched to controls (n = 102) by original trial study arm assignment and ART start date. Additional subsets of HIV infected (n = 41) and HIV uninfected (n = 37) individuals from Improving Recurrence Success (IMPRESS) study were sampled at active TB and post successful treatment completion. Plasma concentrations of soluble adhesion molecules (sMAdCAM, sICAM and sVCAM), lipopolysaccharide binding protein (LBP) and transforming growth factor-beta (TGF-β1, TGF-β2, TGF-β3) were measured by multiplex immunoassays and ELISA. Cytokine data was square root transformed in order to reduce variability. Multivariable analysis adjusted for a number of potential confounders measured at sample time-point: age, BMI, CD4 count, viral load (VL) and measured at baseline: presence or absence of lung cavities, previous history of TB, and WHO disease stage (4 vs 3). The following analytes were associated with increased risk of TB recurrence in the multivariable model: sICAM (aOR 1.06, 95% CI: 1.02-1.12, p = 0.009), LBP (aOR 8.78, 95% CI: 1.23-62.66, p = 0.030) and TGF-β3 (aOR 1.44, 95% CI 1.01-2.05, p = 0.044). Additionally, we observed a positive correlation between LBP and sICAM (r= 0.347, p<0.0001), and LBP and IL-6, identified to be one of the strongest predictors of TB risk in our previous study (r=0.623, p=0.03). These data show that increased risk of TB recurrence in HIV infected individuals on ART is likely associated with HIV mediated translocation of microbial products and the resulting chronic immune activation.
Collapse
Affiliation(s)
- Kimesha Pillay
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Lara Lewis
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Santhuri Rambaran
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Nonhlanhla Yende-Zuma
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | | | - Dhineshree Govender
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Razia Hassan-Moosa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Natasha Samsunder
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.,Department of Epidemiology, Columbia University, New York City, NY, United States
| | - Lyle R McKinnon
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Nesri Padayatchi
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
30
|
Wilkinson KA, Schneider-Luftman D, Lai R, Barrington C, Jhilmeet N, Lowe DM, Kelly G, Wilkinson RJ. Antiretroviral Treatment-Induced Decrease in Immune Activation Contributes to Reduced Susceptibility to Tuberculosis in HIV-1/Mtb Co-infected Persons. Front Immunol 2021; 12:645446. [PMID: 33746987 PMCID: PMC7973093 DOI: 10.3389/fimmu.2021.645446] [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: 12/23/2020] [Accepted: 02/10/2021] [Indexed: 12/27/2022] Open
Abstract
Antiretroviral treatment (ART) reduces the risk of developing active tuberculosis (TB) in HIV-1 co-infected persons. In order to understand host immune responses during ART in the context of Mycobacterium tuberculosis (Mtb) sensitization, we performed RNAseq analysis of whole blood-derived RNA from individuals with latent TB infection coinfected with HIV-1, during the first 6 months of ART. A significant fall in RNA sequence abundance of the Hallmark IFN-alpha, IFN-gamma, IL-6/JAK/STAT3 signaling, and inflammatory response pathway genes indicated reduced immune activation and inflammation at 6 months of ART compared to day 0. Further exploratory evaluation of 65 soluble analytes in plasma confirmed the significant decrease of inflammatory markers after 6 months of ART. Next, we evaluated 30 soluble analytes in QuantiFERON Gold in-tube (QFT) samples from the Ag stimulated and Nil tubes, during the first 6 months of ART in 30 patients. There was a significant decrease in IL-1alpha and IL-1beta (Ag-Nil) concentrations as well as MCP-1 (Nil), supporting decreased immune activation and inflammation. At the same time, IP-10 (Ag-nil) concentrations significantly increased, together with chemokine receptor-expressing CD4 T cell numbers. Our data indicate that ART-induced decrease in immune activation combined with improved antigen responsiveness may contribute to reduced susceptibility to tuberculosis in HIV-1/Mtb co-infected persons.
Collapse
Affiliation(s)
- Katalin A Wilkinson
- Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom.,Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Rachel Lai
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | - Nishtha Jhilmeet
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - David M Lowe
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Gavin Kelly
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, United Kingdom
| | - Robert J Wilkinson
- Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom.,Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
31
|
Esaulova E, Das S, Singh DK, Choreño-Parra JA, Swain A, Arthur L, Rangel-Moreno J, Ahmed M, Singh B, Gupta A, Fernández-López LA, de la Luz Garcia-Hernandez M, Bucsan A, Moodley C, Mehra S, García-Latorre E, Zuniga J, Atkinson J, Kaushal D, Artyomov MN, Khader SA. The immune landscape in tuberculosis reveals populations linked to disease and latency. Cell Host Microbe 2021; 29:165-178.e8. [PMID: 33340449 PMCID: PMC7878437 DOI: 10.1016/j.chom.2020.11.013] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/09/2020] [Accepted: 11/24/2020] [Indexed: 01/06/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) latently infects approximately one-fourth of the world's population. The immune mechanisms that govern progression from latent (LTBI) to active pulmonary TB (PTB) remain poorly defined. Experimentally Mtb-infected non-human primates (NHP) mirror the disease observed in humans and recapitulate both PTB and LTBI. We characterized the lung immune landscape in NHPs with LTBI and PTB using high-throughput technologies. Three defining features of PTB in macaque lungs include the influx of plasmacytoid dendritic cells (pDCs), an Interferon (IFN)-responsive macrophage population, and activated T cell responses. In contrast, a CD27+ Natural killer (NK) cell subset accumulated in the lungs of LTBI macaques. This NK cell population was also detected in the circulation of LTBI individuals. This comprehensive analysis of the lung immune landscape will improve the understanding of TB immunopathogenesis, providing potential targets for therapies and vaccines for TB control.
Collapse
Affiliation(s)
- Ekaterina Esaulova
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Shibali Das
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dhiraj Kumar Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Jose Alberto Choreño-Parra
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City 14080, Mexico; Laboratorio de Inmunoquímica I, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - Amanda Swain
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Laura Arthur
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Javier Rangel-Moreno
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Mushtaq Ahmed
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bindu Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Ananya Gupta
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Luis Alejandro Fernández-López
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City 14080, Mexico; Laboratorio de Inmunoquímica I, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - Maria de la Luz Garcia-Hernandez
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Allison Bucsan
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington 70112, Louisiana
| | - Chivonne Moodley
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington 70112, Louisiana
| | - Smriti Mehra
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington 70112, Louisiana
| | - Ethel García-Latorre
- Laboratorio de Inmunoquímica I, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - Joaquin Zuniga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City 14080, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City 07320 Mexico
| | - Jeffrey Atkinson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA.
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
32
|
Early innate and adaptive immune perturbations determine long-term severity of chronic virus and Mycobacterium tuberculosis coinfection. Immunity 2021; 54:526-541.e7. [PMID: 33515487 DOI: 10.1016/j.immuni.2021.01.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/13/2020] [Accepted: 01/11/2021] [Indexed: 01/01/2023]
Abstract
Chronic viral infections increase severity of Mycobacterium tuberculosis (Mtb) coinfection. Here, we examined how chronic viral infections alter the pulmonary microenvironment to foster coinfection and worsen disease severity. We developed a coordinated system of chronic virus and Mtb infection that induced central clinical manifestations of coinfection, including increased Mtb burden, extra-pulmonary dissemination, and heightened mortality. These disease states were not due to chronic virus-induced immunosuppression or exhaustion; rather, increased amounts of the cytokine TNFα initially arrested pulmonary Mtb growth, impeding dendritic cell mediated antigen transportation to the lymph node and subverting immune-surveillance, allowing bacterial sanctuary. The cryptic Mtb replication delayed CD4 T cell priming, redirecting T helper (Th) 1 toward Th17 differentiation and increasing pulmonary neutrophilia, which diminished long-term survival. Temporally restoring CD4 T cell induction overcame these diverse disease sequelae to enhance Mtb control. Thus, Mtb co-opts TNFα from the chronic inflammatory environment to subvert immune-surveillance, avert early immune function, and foster long-term coinfection.
Collapse
|
33
|
Al-Mozaini M, Alrahbeni T, Dirar Q, Alotibi J, Alrajhi A. HIV in the Kingdom of Saudi Arabia: Can We Change the Way We Deal with Co-Infections. Infect Drug Resist 2021; 14:111-117. [PMID: 33500633 PMCID: PMC7822075 DOI: 10.2147/idr.s270355] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022] Open
Abstract
The first incidence of acquired immunodeficiency syndrome (AIDS) from the Kingdom of Saudi Arabia (KSA) was reported back in 1984, and by the end of 2013, around 1509 patients were diagnosed with HIV infection. Recently in 2018, the Saudi ministry of health released that the incidence of HIV in Saudi Arabia is 3 cases of HIV for every 10,000 of the population. Having said that, the surveillance of HIV will face a range of challenges in KSA despite proper medical care, counseling, family planning, diagnostic, evaluation, and the use of effective anti-retroviral therapy. Patients who underwent anti-retroviral therapy showed significant reduction in morbidity as well as mortality. On the other hand, further targeted treatment and preventive strategies are warranted to control HIV co-infections in the KSA. In addition, progress towards meeting the WHO 90-90-90 goals for HIV not only at KSA but at the MENA region too, which is that of the population, 90% are diagnosed, 90% undergoing treatment, and 90% under viral control, is not being systematically monitored. In this review, we discuss the common co-infections with HIV infections that are reported in KSA, which when compared to international trends, it is similar for both viral hepatitis and tuberculosis. Although those co-infections exist, they are presented in different ratios and percentages when compared to the international reported data. These differences mandates defining and introducing new resilient methods of treatment and preventive measures. In this review, we offer an insight into healthcare policymakers to be compliant with UNAIDS 2020 vision program. We also discuss some of the gaps and recommendations to achieve the WHO 90-90-90 goal.
Collapse
Affiliation(s)
- Maha Al-Mozaini
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Qais Dirar
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Jawaher Alotibi
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Abdulrahman Alrajhi
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Singh DK, Singh B, Ganatra SR, Gazi M, Cole J, Thippeshappa R, Alfson KJ, Clemmons E, Gonzalez O, Escobedo R, Lee TH, Chatterjee A, Goez-Gazi Y, Sharan R, Gough M, Alvarez C, Blakley A, Ferdin J, Bartley C, Staples H, Parodi L, Callery J, Mannino A, Klaffke B, Escareno P, Platt RN, Hodara V, Scordo J, Gautam S, Vilanova AG, Olmo-Fontanez A, Schami A, Oyejide A, Ajithdoss DK, Copin R, Baum A, Kyratsous C, Alvarez X, Ahmed M, Rosa B, Goodroe A, Dutton J, Hall-Ursone S, Frost PA, Voges AK, Ross CN, Sayers K, Chen C, Hallam C, Khader SA, Mitreva M, Anderson TJC, Martinez-Sobrido L, Patterson JL, Turner J, Torrelles JB, Dick EJ, Brasky K, Schlesinger LS, Giavedoni LD, Carrion R, Kaushal D. Responses to acute infection with SARS-CoV-2 in the lungs of rhesus macaques, baboons and marmosets. Nat Microbiol 2021; 6:73-86. [PMID: 33340034 PMCID: PMC7890948 DOI: 10.1038/s41564-020-00841-4] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022]
Abstract
Non-human primate models will expedite therapeutics and vaccines for coronavirus disease 2019 (COVID-19) to clinical trials. Here, we compare acute severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in young and old rhesus macaques, baboons and old marmosets. Macaques had clinical signs of viral infection, mild to moderate pneumonitis and extra-pulmonary pathologies, and both age groups recovered in two weeks. Baboons had prolonged viral RNA shedding and substantially more lung inflammation compared with macaques. Inflammation in bronchoalveolar lavage was increased in old versus young baboons. Using techniques including computed tomography imaging, immunophenotyping, and alveolar/peripheral cytokine response and immunohistochemical analyses, we delineated cellular immune responses to SARS-CoV-2 infection in macaque and baboon lungs, including innate and adaptive immune cells and a prominent type-I interferon response. Macaques developed T-cell memory phenotypes/responses and bystander cytokine production. Old macaques had lower titres of SARS-CoV-2-specific IgG antibody levels compared with young macaques. Acute respiratory distress in macaques and baboons recapitulates the progression of COVID-19 in humans, making them suitable as models to test vaccines and therapies.
Collapse
Affiliation(s)
- Dhiraj Kumar Singh
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Bindu Singh
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Shashank R Ganatra
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Michal Gazi
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Journey Cole
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Rajesh Thippeshappa
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Elizabeth Clemmons
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Olga Gonzalez
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ruby Escobedo
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Tae-Hyung Lee
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ayan Chatterjee
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Riti Sharan
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Maya Gough
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Cynthia Alvarez
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Alyssa Blakley
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Justin Ferdin
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Carmen Bartley
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Hilary Staples
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Laura Parodi
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jessica Callery
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Amanda Mannino
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | | | - Roy N Platt
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Vida Hodara
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Julia Scordo
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Shalini Gautam
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | | | - Alyssa Schami
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | | | | | - Alina Baum
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | | | - Xavier Alvarez
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Mushtaq Ahmed
- Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Bruce Rosa
- Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Anna Goodroe
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - John Dutton
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Shannan Hall-Ursone
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Patrice A Frost
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Andra K Voges
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
- Veterinary Imaging Consulting of South Texas, San Antonio, TX, USA
| | - Corinna N Ross
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ken Sayers
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Christopher Chen
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Cory Hallam
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Shabaana A Khader
- Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Makedonka Mitreva
- Washington University School of Medicine in St Louis, St Louis, MO, USA
| | | | | | | | - Joanne Turner
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Edward J Dick
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Kathleen Brasky
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Larry S Schlesinger
- Southwest National Primate Research Center, San Antonio, TX, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Luis D Giavedoni
- Southwest National Primate Research Center, San Antonio, TX, USA.
- Texas Biomedical Research Institute, San Antonio, TX, USA.
| | - Ricardo Carrion
- Southwest National Primate Research Center, San Antonio, TX, USA.
- Texas Biomedical Research Institute, San Antonio, TX, USA.
| | - Deepak Kaushal
- Southwest National Primate Research Center, San Antonio, TX, USA.
- Texas Biomedical Research Institute, San Antonio, TX, USA.
| |
Collapse
|
35
|
Rakshit S, Hingankar N, Alampalli SV, Adiga V, Sundararaj BK, Sahoo PN, Finak G, Uday Kumar J AJ, Dhar C, D'Souza G, Virkar RG, Ghate M, Thakar MR, Paranjape RS, De Rosa SC, Ottenhoff THM, Vyakarnam A. HIV Skews a Balanced Mtb-Specific Th17 Response in Latent Tuberculosis Subjects to a Pro-inflammatory Profile Independent of Viral Load. Cell Rep 2020; 33:108451. [PMID: 33264614 DOI: 10.1016/j.celrep.2020.108451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 09/15/2020] [Accepted: 11/09/2020] [Indexed: 02/08/2023] Open
Abstract
HIV infection predisposes latent tuberculosis-infected (LTBI) subjects to active TB. This study is designed to determine whether HIV infection of LTBI subjects compromises the balanced Mycobacterium tuberculosis (Mtb)-specific T helper 17 (Th17) response of recognized importance in anti-TB immunity. Comparative analysis of Mtb- and cytomegalovirus (CMV)-specific CD4+ T cell responses demonstrates a marked dampening of the Mtb-specific CD4+ T cell effectors and polyfunctional cells while preserving CMV-specific response. Additionally, HIV skews the Mtb-specific Th17 response in chronic HIV-infected LTBI progressors, but not long-term non-progressors (LTNPs), with preservation of pro-inflammatory interferon (IFN)-γ+/interleukin-17+ (IL-17+) and significant loss of anti-inflammatory IL-10+/IL-17+ effectors that is restored by anti-retroviral therapy (ART). HIV-driven impairment of Mtb-specific response cannot be attributed to preferential infection as cell-associated HIV DNA and HIV RNA reveal equivalent viral burden in CD4+ T cells from different antigen specificities. We therefore propose that beyond HIV-induced loss of Mtb-specific CD4+ T cells, the associated dysregulation of Mtb-specific T cell homeostasis can potentially enhance the onset of TB in LTBI subjects.
Collapse
Affiliation(s)
- Srabanti Rakshit
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Nitin Hingankar
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Shuba Varshini Alampalli
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Bharath K Sundararaj
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Pravat Nalini Sahoo
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Anto Jesuraj Uday Kumar J
- Departments of Infectious Diseases & Pulmonary Medicine, St. John's Research Institute, Bangalore, India
| | - Chirag Dhar
- Departments of Infectious Diseases & Pulmonary Medicine, St. John's Research Institute, Bangalore, India
| | - George D'Souza
- Departments of Infectious Diseases & Pulmonary Medicine, St. John's Research Institute, Bangalore, India
| | | | - Manisha Ghate
- National Aids Research Institute, Bhosari, Pune, Maharashtra, India
| | - Madhuri R Thakar
- National Aids Research Institute, Bhosari, Pune, Maharashtra, India
| | | | - Stephen C De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, Guy's Hospital, King's College London, London SE1 9RT, UK.
| |
Collapse
|
36
|
Weinberg A, Tugizov S, Pandiyan P, Jin G, Rakshit S, Vyakarnam A, Naglik JR. Innate immune mechanisms to oral pathogens in oral mucosa of HIV-infected individuals. Oral Dis 2020; 26 Suppl 1:69-79. [PMID: 32862519 PMCID: PMC7570967 DOI: 10.1111/odi.13470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A crucial aspect of mucosal HIV transmission is the interaction between HIV, the local environmental milieu and immune cells. The oral mucosa comprises many host cell types including epithelial cells, CD4 + T cells, dendritic cells and monocytes/macrophages, as well as a diverse microbiome predominantly comprising bacterial species. While the oral epithelium is one of the first sites exposed to HIV through oral-genital contact and nursing infants, it is largely thought to be resistant to HIV transmission via mechanisms that are still unclear. HIV-1 infection is also associated with predisposition to secondary infections, such as tuberculosis, and other diseases including cancer. This review addresses the following questions that were discussed at the 8th World Workshop on Oral Health and Disease in AIDS held in Bali, Indonesia, 13 September –15 September 2019: (a) How does HIV infection affect epithelial cell signalling? (b) How does HIV infection affect the production of cytokines and other innate antimicrobial factors, (c) How is the mucosal distribution and function of immune cells altered in HIV infection? (d) How do T cells affect HIV (oral) pathogenesis and cancer? (e) How does HIV infection lead to susceptibility to TB infections?
Collapse
Affiliation(s)
- Aaron Weinberg
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sharof Tugizov
- Department of Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ge Jin
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Srabanti Rakshit
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Annapurna Vyakarnam
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
37
|
Ivanyi J. Tuberculosis vaccination needs to avoid 'decoy' immune reactions. Tuberculosis (Edinb) 2020; 126:102021. [PMID: 33254012 DOI: 10.1016/j.tube.2020.102021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 11/16/2022]
Abstract
Current search for a new effective vaccine against tuberculosis involves selected antigens, vectors and adjuvants. These are being evaluated usually by their booster inoculation following priming with Bacillus Calmette-Guerin. The purpose of this article is to point out, that despite being attenuated of virulence, priming with BCG may still involve immune mechanisms, which are not favourable for protection against active disease. It is postulated, that the responsible 'decoy' constituents selected during the evolution of pathogenic tubercle bacilli may be involved in the evasion from bactericidal host resistance and stimulate immune responses of a cytokine phenotype, which lead to the transition from latent closed granulomas to reactivation with infectious lung cavities. The decoy mechanisms appear as favourable for most infected subjects but leading in a minority of cases to pathology which can effectively transmit the infection. It is proposed that construction and development of new vaccine candidates could benefit from avoiding decoy-type immune mechanisms.
Collapse
Affiliation(s)
- Juraj Ivanyi
- Centre for Host-Microbiome Interactions, Guy's Campus of Kings College London, SE1, 1UL, United kingdom.
| |
Collapse
|
38
|
Sharan R, Kaushal D. Vaccine strategies for the Mtb/HIV copandemic. NPJ Vaccines 2020; 5:95. [PMID: 33083030 PMCID: PMC7555484 DOI: 10.1038/s41541-020-00245-9] [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: 06/06/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
One-third of world’s population is predicted to be infected with tuberculosis (TB). The resurgence of this deadly disease has been inflamed by comorbidity with human immunodeficiency virus (HIV). The risk of TB in people living with HIV (PLWH) is 15–22 times higher than people without HIV. Development of a single vaccine to combat both diseases is an ardent but tenable ambition. Studies have focused on the induction of specific humoral and cellular immune responses against HIV-1 following recombinant BCG (rBCG) expressing HIV-1 antigens. Recent advances in the TB vaccines led to the development of promising candidates such as MTBVAC, the BCG revaccination approach, H4:IC31, H56:IC31, M72/AS01 and more recently, intravenous (IV) BCG. Modification of these vaccine candidates against TB/HIV coinfection could reveal key correlates of protection in a representative animal model. This review discusses the (i) potential TB vaccine candidates that can be exploited for use as a dual vaccine against TB/HIV copandemic (ii) progress made in the realm of TB/HIV dual vaccine candidates in small animal model, NHP model, and human clinical trials (iii) the failures and promising targets for a successful vaccine strategy while delineating the correlates of vaccine-induced protection.
Collapse
Affiliation(s)
- Riti Sharan
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX 78227 USA
| | - Deepak Kaushal
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX 78227 USA
| |
Collapse
|
39
|
Ganatra SR, Bucşan AN, Alvarez X, Kumar S, Chatterjee A, Quezada M, Fish A, Singh DK, Singh B, Sharan R, Lee TH, Shanmugasundaram U, Velu V, Khader SA, Mehra S, Rengarajan J, Kaushal D. Antiretroviral therapy does not reduce tuberculosis reactivation in a tuberculosis-HIV coinfection model. J Clin Invest 2020; 130:5171-5179. [PMID: 32544085 PMCID: PMC7524506 DOI: 10.1172/jci136502] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
While the advent of combination antiretroviral therapy (ART) has significantly improved survival, tuberculosis (TB) remains the leading cause of death in the HIV-infected population. We used Mycobacterium tuberculosis/simian immunodeficiency virus-coinfected (M. tuberculosis/SIV-coinfected) macaques to model M. tuberculosis/HIV coinfection and study the impact of ART on TB reactivation due to HIV infection. Although ART significantly reduced viral loads and increased CD4+ T cell counts in blood and bronchoalveolar lavage (BAL) samples, it did not reduce the relative risk of SIV-induced TB reactivation in ART-treated macaques in the early phase of treatment. CD4+ T cells were poorly restored specifically in the lung interstitium, despite their significant restoration in the alveolar compartment of the lung as well as in the periphery. IDO1 induction in myeloid cells in the inducible bronchus-associated lymphoid tissue (iBALT) likely contributed to dysregulated T cell homing and impaired lung immunity. Thus, although ART was indispensable for controlling viral replication, restoring CD4+ T cells, and preventing opportunistic infection, it appeared inadequate in reversing the clinical signs of TB reactivation during the relatively short duration of ART administered in this study. This finding warrants the modeling of concurrent treatment of TB and HIV to potentially reduce the risk of reactivation of TB due to HIV to inform treatment strategies in patients with M. tuberculosis/HIV coinfection.
Collapse
Affiliation(s)
- Shashank R. Ganatra
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Allison N. Bucşan
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Xavier Alvarez
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Shyamesh Kumar
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Ayan Chatterjee
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Melanie Quezada
- Emory Vaccine Center and
- Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Abigail Fish
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Dhiraj K. Singh
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Bindu Singh
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Riti Sharan
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Tae-Hyung Lee
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Uma Shanmugasundaram
- Emory Vaccine Center and
- Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vijayakumar Velu
- Emory Vaccine Center and
- Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Smriti Mehra
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Jyothi Rengarajan
- Emory Vaccine Center and
- Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
40
|
Sharan R, Bucşan AN, Ganatra S, Paiardini M, Mohan M, Mehra S, Khader SA, Kaushal D. Chronic Immune Activation in TB/HIV Co-infection. Trends Microbiol 2020; 28:619-632. [PMID: 32417227 PMCID: PMC7390597 DOI: 10.1016/j.tim.2020.03.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/03/2020] [Accepted: 03/25/2020] [Indexed: 12/27/2022]
Abstract
HIV co-infection is the most critical risk factor for the reactivation of latent tuberculosis (TB) infection (LTBI). While CD4+ T cell depletion has been considered the major cause of HIV-induced reactivation of LTBI, recent work in macaques co-infected with Mycobacterium tuberculosis (Mtb)/simian immunodeficiency virus (SIV) suggests that cytopathic effects of SIV resulting in chronic immune activation and dysregulation of T cell homeostasis correlate with reactivation of LTBI. This review builds on compelling data that the reactivation of LTBI during HIV co-infection is likely to be driven by the events of HIV replication and therefore highlights the need to have optimum translational interventions directed at reactivation due to co-infection.
Collapse
Affiliation(s)
- Riti Sharan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Allison N Bucşan
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Shashank Ganatra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Mirko Paiardini
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Mahesh Mohan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| |
Collapse
|
41
|
Diedrich CR, Rutledge T, Maiello P, Baranowski TM, White AG, Borish HJ, Karell P, Hopkins F, Brown J, Fortune SM, Flynn JL, Ambrose Z, Lin PL. SIV and Mycobacterium tuberculosis synergy within the granuloma accelerates the reactivation pattern of latent tuberculosis. PLoS Pathog 2020; 16:e1008413. [PMID: 32730321 PMCID: PMC7419014 DOI: 10.1371/journal.ppat.1008413] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 08/11/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022] Open
Abstract
Human immunodeficiency virus infection is the most common risk factor for severe forms of tuberculosis (TB), regardless of CD4 T cell count. Using a well-characterized cynomolgus macaque model of human TB, we compared radiographic, immunologic and microbiologic characteristics of early (subclinical) reactivation of latent M. tuberculosis (Mtb) infection among animals subsequently infected with simian immunodeficiency virus (SIV) or who underwent anti-CD4 depletion by a depletion antibody. CD4 depleted animals had significantly fewer CD4 T cells within granulomas compared to Mtb/SIV co-infected and Mtb-only control animals. After 2 months of treatment, subclinical reactivation occurred at similar rates among CD4 depleted (5 of 7 animals) and SIV infected animals (4 of 8 animals). However, SIV-induced reactivation was associated with more dissemination of lung granulomas that were permissive to Mtb growth resulting in greater bacterial burden within granulomas compared to CD4 depleted reactivators. Granulomas from Mtb/SIV animals displayed a more robust T cell activation profile (IFN-α, IFN-γ, TNF, IL-17, IL-2, IL-10, IL-4 and granzyme B) compared to CD4 depleted animals and controls though these effectors did not protect against reactivation or dissemination, but instead may be related to increased viral and/or Mtb antigens. SIV replication within the granuloma was associated with reactivation, greater overall Mtb growth and reduced Mtb killing resulting in greater overall Mtb burden. These data support that SIV disrupts protective immune responses against latent Mtb infection beyond the loss of CD4 T cells, and that synergy between SIV and Mtb occurs within granulomas.
Collapse
Affiliation(s)
- Collin R. Diedrich
- Department of Pediatrics, Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Tara Rutledge
- Department of Pediatrics, Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Pauline Maiello
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Tonilynn M. Baranowski
- Department of Pediatrics, Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Alexander G. White
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - H. Jacob Borish
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Paul Karell
- Department of Pediatrics, Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Forrest Hopkins
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jessica Brown
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - JoAnne L. Flynn
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Zandrea Ambrose
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Philana Ling Lin
- Department of Pediatrics, Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
42
|
Shanmugasundaram U, Bucsan AN, Ganatra SR, Ibegbu C, Quezada M, Blair RV, Alvarez X, Velu V, Kaushal D, Rengarajan J. Pulmonary Mycobacterium tuberculosis control associates with CXCR3- and CCR6-expressing antigen-specific Th1 and Th17 cell recruitment. JCI Insight 2020; 5:137858. [PMID: 32554933 DOI: 10.1172/jci.insight.137858] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/04/2020] [Indexed: 12/15/2022] Open
Abstract
Mycobacterium tuberculosis-specific (M. tuberculosis-specific) T cell responses associated with immune control during asymptomatic latent tuberculosis infection (LTBI) remain poorly understood. Using a nonhuman primate aerosol model, we studied the kinetics, phenotypes, and functions of M. tuberculosis antigen-specific T cells in peripheral and lung compartments of M. tuberculosis-infected asymptomatic rhesus macaques by longitudinally sampling blood and bronchoalveolar lavage, for up to 24 weeks postinfection. We found substantially higher frequencies of M. tuberculosis-specific effector and memory CD4+ and CD8+ T cells producing IFN-γ in the airways compared with peripheral blood, and these frequencies were maintained throughout the study period. Moreover, M. tuberculosis-specific IL-17+ and IL-17+IFN-γ+ double-positive T cells were present in the airways but were largely absent in the periphery, suggesting that balanced mucosal Th1/Th17 responses are associated with LTBI. The majority of M. tuberculosis-specific CD4+ T cells that homed to the airways expressed the chemokine receptor CXCR3 and coexpressed CCR6. Notably, CXCR3+CD4+ cells were found in granulomatous and nongranulomatous regions of the lung and inversely correlated with M. tuberculosis burden. Our findings provide insights into antigen-specific T cell responses associated with asymptomatic M. tuberculosis infection that are relevant for developing better strategies to control TB.
Collapse
Affiliation(s)
| | - Allison N Bucsan
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
| | - Shashank R Ganatra
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA.,Southwest National Primate Research Center, San Antonio, Texas, USA.,Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Chris Ibegbu
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Melanie Quezada
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
| | - Robert V Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Xavier Alvarez
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA.,Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Vijayakumar Velu
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Deepak Kaushal
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA.,Southwest National Primate Research Center, San Antonio, Texas, USA.,Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
43
|
Thippeshappa R, Kimata JT, Kaushal D. Toward a Macaque Model of HIV-1 Infection: Roadblocks, Progress, and Future Strategies. Front Microbiol 2020; 11:882. [PMID: 32477302 PMCID: PMC7237640 DOI: 10.3389/fmicb.2020.00882] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022] Open
Abstract
The human-specific tropism of Human Immunodeficiency Virus Type 1 (HIV-1) has complicated the development of a macaque model of HIV-1 infection/AIDS that is suitable for preclinical evaluation of vaccines and novel treatment strategies. Several innate retroviral restriction factors, such as APOBEC3 family of proteins, TRIM5α, BST2, and SAMHD1, that prevent HIV-1 replication have been identified in macaque cells. Accessory proteins expressed by Simian Immunodeficiency virus (SIV) such as viral infectivity factor (Vif), viral protein X (Vpx), viral protein R (Vpr), and negative factor (Nef) have been shown to play key roles in overcoming these restriction factors in macaque cells. Thus, substituting HIV-1 accessory genes with those from SIV may enable HIV-1 replication in macaques. We and others have constructed macaque-tropic HIV-1 derivatives [also called simian-tropic HIV-1 (stHIV-1) or Human-Simian Immunodeficiency Virus (HSIV)] carrying SIV vif to overcome APOBEC3 family proteins. Additional modifications to HIV-1 gag in some of the macaque-tropic HIV-1 have also been done to overcome TRIM5α restriction in rhesus and cynomolgus macaques. Although these viruses replicate persistently in macaque species, they do not result in CD4 depletion. Thus, these studies suggest that additional blocks to HIV-1 replication exist in macaques that prevent high-level viral replication. Furthermore, serial animal-to-animal passaging of macaque-tropic HIV-1 in vivo has not resulted in pathogenic variants that cause AIDS in immunocompetent macaques. In this review, we discuss recent developments made toward developing macaque model of HIV-1 infection.
Collapse
Affiliation(s)
- Rajesh Thippeshappa
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
44
|
Ellis AL, Balgeman AJ, Larson EC, Rodgers MA, Ameel C, Baranowski T, Kannal N, Maiello P, Juno JA, Scanga CA, O’Connor SL. MAIT cells are functionally impaired in a Mauritian cynomolgus macaque model of SIV and Mtb co-infection. PLoS Pathog 2020; 16:e1008585. [PMID: 32433713 PMCID: PMC7266356 DOI: 10.1371/journal.ppat.1008585] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/02/2020] [Accepted: 04/29/2020] [Indexed: 12/11/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells can recognize and respond to some bacterially infected cells. Several in vitro and in vivo models of Mycobacterium tuberculosis (Mtb) infection suggest that MAIT cells can contribute to control of Mtb, but these studies are often cross-sectional and use peripheral blood cells. Whether MAIT cells are recruited to Mtb-affected granulomas and lymph nodes (LNs) during early Mtb infection and what purpose they might serve there is less well understood. Furthermore, whether HIV/SIV infection impairs MAIT cell frequency or function at the sites of Mtb replication has not been determined. Using Mauritian cynomolgus macaques (MCM), we phenotyped MAIT cells in the peripheral blood and bronchoalveolar lavage (BAL) before and during infection with SIVmac239. To test the hypothesis that SIV co-infection impairs MAIT cell frequency and function within granulomas, SIV+ and -naïve MCM were infected with a low dose of Mtb Erdman, and necropsied at 6 weeks post Mtb-challenge. MAIT cell frequency and function were examined within the peripheral blood, BAL, and Mtb-affected lymph nodes (LN) and granulomas. MAIT cells did not express markers indicative of T cell activation in response to Mtb in vivo within granulomas in animals infected with Mtb alone. SIV and Mtb co-infection led to increased expression of the activation/exhaustion markers PD-1 and TIGIT, and decreased ability to secrete TNFα when compared to SIV-naïve MCM. Our study provides evidence that SIV infection does not prohibit the recruitment of MAIT cells to sites of Mtb infection, but does functionally impair those MAIT cells. Their impaired function could have impacts, either direct or indirect, on the long-term containment of TB disease.
Collapse
Affiliation(s)
- Amy L. Ellis
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Alexis J. Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Erica C. Larson
- Department of Microbiology and Molecular Genetics, and Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mark A. Rodgers
- Department of Microbiology and Molecular Genetics, and Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Cassaundra Ameel
- Department of Microbiology and Molecular Genetics, and Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Tonilynn Baranowski
- Department of Microbiology and Molecular Genetics, and Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Nadean Kannal
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, and Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, and Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
45
|
McLean MR, Lu LL, Kent SJ, Chung AW. An Inflammatory Story: Antibodies in Tuberculosis Comorbidities. Front Immunol 2019; 10:2846. [PMID: 31921122 PMCID: PMC6913197 DOI: 10.3389/fimmu.2019.02846] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/19/2019] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) resides in a quarter of the world's population and is the causative agent for tuberculosis (TB), the most common infectious reason of death in humans today. Although cellular immunity has been firmly established in the control of Mtb, there is growing evidence that antibodies may also modulate the infection. More specifically, certain antibody features are associated with inflammation and are divergent in different states of human infection and disease. Importantly, TB impacts not just the healthy but also those with chronic conditions. While HIV represents the quintessential comorbid condition for TB, recent epidemiological evidence shows that additional chronic conditions such as diabetes and kidney disease are rising. In fact, the prevalence of diabetes as a comorbid TB condition is now higher than that of HIV. These chronic diseases are themselves independently associated with pro-inflammatory immune states that encompass antibody profiles. This review discusses isotypes, subclasses, post-translational modifications and Fc-mediated functions of antibodies in TB infection and in the comorbid chronic conditions of HIV, diabetes, and kidney diseases. We propose that inflammatory antibody profiles, which are a marker of active TB, may be an important biomarker for detection of TB disease progression within comorbid individuals. We highlight the need for future studies to determine which inflammatory antibody profiles are the consequences of comorbidities and which may potentially contribute to TB reactivation.
Collapse
Affiliation(s)
- Milla R McLean
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Lenette L Lu
- Division of Infectious Disease and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Infectious Diseases Department, Melbourne Sexual Health Centre, Alfred Health, Central Clinical School, Monash University, Brisbane, VIC, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, SA, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
46
|
Affiliation(s)
- Sophia Häfner
- University of Copenhagen, BRIC Biotech Research & Innovation Centre, Lund Group, 2200 Copenhagen, Denmark.
| |
Collapse
|