1
|
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
|
2
|
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
|
3
|
Bohórquez JA, Adduri S, Ansari D, John S, Florence J, Adejare O, Singh G, Konduru NV, Jagannath C, Yi G. A novel humanized mouse model for HIV and tuberculosis co-infection studies. Front Immunol 2024; 15:1395018. [PMID: 38799434 PMCID: PMC11116656 DOI: 10.3389/fimmu.2024.1395018] [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: 03/02/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Background Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), continues to be a major public health problem worldwide. The human immunodeficiency virus (HIV) is another equally important life-threatening pathogen. HIV infection decreases CD4+ T cell levels markedly increasing Mtb co-infections. An appropriate animal model for HIV/Mtb co-infection that can recapitulate the diversity of the immune response in humans during co-infection would facilitate basic and translational research in HIV/Mtb infections. Herein, we describe a novel humanized mouse model. Methods The irradiated NSG-SGM3 mice were transplanted with human CD34+ hematopoietic stem cells, and the humanization was monitored by staining various immune cell markers for flow cytometry. They were challenged with HIV and/or Mtb, and the CD4+ T cell depletion and HIV viral load were monitored over time. Before necropsy, the live mice were subjected to pulmonary function test and CT scan, and after sacrifice, the lung and spleen homogenates were used to determine Mtb load (CFU) and cytokine/chemokine levels by multiplex assay, and lung sections were analyzed for histopathology. The mouse sera were subjected to metabolomics analysis. Results Our humanized NSG-SGM3 mice were able to engraft human CD34+ stem cells, which then differentiated into a full-lineage of human immune cell subsets. After co-infection with HIV and Mtb, these mice showed decrease in CD4+ T cell counts overtime and elevated HIV load in the sera, similar to the infection pattern of humans. Additionally, Mtb caused infections in both lungs and spleen, and induced granulomatous lesions in the lungs. Distinct metabolomic profiles were also observed in the tissues from different mouse groups after co-infections. Conclusion The humanized NSG-SGM3 mice are able to recapitulate the pathogenic effects of HIV and Mtb infections and co-infection at the pathological, immunological and metabolism levels and are therefore a reproducible small animal model for studying HIV/Mtb co-infection.
Collapse
Affiliation(s)
- José Alejandro Bohórquez
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX, United States
| | - Sitaramaraju Adduri
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Danish Ansari
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX, United States
| | - Sahana John
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX, United States
| | - Jon Florence
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Omoyeni Adejare
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Gaurav Singh
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX, United States
| | - Nagarjun V. Konduru
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Center for Infectious Diseases and Translational Medicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Guohua Yi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX, United States
| |
Collapse
|
4
|
Bohórquez JA, Adduri S, Ansari D, John S, Florence J, Adejare O, Singh G, Konduru N, Jagannath C, Yi G. A Novel Humanized Mouse Model for HIV and Tuberculosis Co-infection Studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583545. [PMID: 38496484 PMCID: PMC10942347 DOI: 10.1101/2024.03.05.583545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), continues to be a major public health problem worldwide. The human immunodeficiency virus (HIV) is another equally important life-threatening pathogen. Further, co-infections with HIV and Mtb have severe effects in the host, with people infected with HIV being fifteen to twenty-one times more likely to develop active TB. The use of an appropriate animal model for HIV/Mtb co-infection that can recapitulate the diversity of the immune response in humans would be a useful tool for conducting basic and translational research in HIV/Mtb infections. The present study was focused on developing a humanized mouse model for investigations on HIV-Mtb co-infection. Using NSG-SGM3 mice that can engraft human stem cells, our studies showed that they were able to engraft human CD34+ stem cells which then differentiate into a full-lineage of human immune cell subsets. After co-infection with HIV and Mtb, these mice showed decrease in CD4+ T cell counts overtime and elevated HIV load in the sera, similar to the infection pattern of humans. Additionally, Mtb caused infections in both lungs and spleen, and induced the development of granulomatous lesions in the lungs, detected by CT scan and histopathology. Distinct metabolomic profiles were also observed in the tissues from different mouse groups after co-infections. Our results suggest that the humanized NSG-SGM3 mice are able to recapitulate the effects of HIV and Mtb infections and co-infection in the human host at pathological, immunological and metabolism levels, providing a dependable small animal model for studying HIV/Mtb co-infection.
Collapse
Affiliation(s)
- José Alejandro Bohórquez
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
| | - Sitaramaraju Adduri
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Danish Ansari
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
| | - Sahana John
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
| | - Jon Florence
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Omoyeni Adejare
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Gaurav Singh
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
| | - Nagarjun Konduru
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Center for Infectious Diseases and Translational Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Guohua Yi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
| |
Collapse
|
5
|
Adeyemo S, Sangotola A, Korosteleva O. Modeling Transmission Dynamics of Tuberculosis-HIV Co-Infection in South Africa. EPIDEMIOLOGIA 2023; 4:408-419. [PMID: 37873885 PMCID: PMC10594517 DOI: 10.3390/epidemiologia4040036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
South Africa has the highest number of people living with the human immunodeficiency virus (HIV) in the world, accounting for nearly one in five people living with HIV globally. As of 2021, 8 million people in South Africa were infected with HIV, which is 13% of the country's total population. Approximately 450,000 people in the country develop tuberculosis (TB) disease every year, and 270,000 of those are HIV positive. This suggests that being HIV positive significantly increases one's susceptibility to TB, accelerating the spread of the epidemic. To better understand the disease burden at the population level, a Susceptible-Infected-Recovered-Dead (SIRD) TB-HIV co-infection epidemic model is presented. Parameter values are estimated using the method of moments. The disease-free equilibrium and basic reproduction number of the model are also obtained. Finally, numeric simulations are carried out for a 30-year period to give insights into the transmission dynamics of the co-infection.
Collapse
Affiliation(s)
- Simeon Adeyemo
- Department of Mathematics and Statistics, California State University, Long Beach, CA 90840, USA;
| | - Adekunle Sangotola
- Department of Physical Sciences, Bells University of Technology, Ota 112212, Ogun, Nigeria;
| | - Olga Korosteleva
- Department of Mathematics and Statistics, California State University, Long Beach, CA 90840, USA;
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Clain JA, Rabezanahary H, Racine G, Boutrais S, Soundaramourty C, Joly Beauparlant C, Jenabian MA, Droit A, Ancuta P, Zghidi-Abouzid O, Estaquier J. Early ART reduces viral seeding and innate immunity in liver and lungs of SIV-infected macaques. JCI Insight 2023; 8:e167856. [PMID: 37485876 PMCID: PMC10443800 DOI: 10.1172/jci.insight.167856] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Identifying immune cells and anatomical tissues that contribute to the establishment of viral reservoirs is of central importance in HIV-1 cure research. Herein, we used rhesus macaques (RMs) infected with SIVmac251 to analyze viral seeding in the liver and lungs of either untreated or early antiretroviral therapy-treated (ART-treated) RMs. Consistent with viral replication and sensing, transcriptomic analyses showed higher levels of inflammation, pyroptosis, and chemokine genes as well as of interferon-stimulating gene (ISG) transcripts, in the absence of ART. Our results highlighted the infiltration of monocyte-derived macrophages (HLA-DR+CD11b+CD14+CD16+) in inflamed liver and lung tissues associated with the expression of CD183 and CX3CR1 but also with markers of tissue-resident macrophages (CD206+ and LYVE+). Sorting of myeloid cell subsets demonstrated that CD14+CD206-, CD14+CD206+, and CD14-CD206+ cell populations were infected, in the liver and lungs, in SIVmac251-infected RMs. Of importance, early ART drastically reduced viral seeding consistent with the absence of ISG detection but also of genes related to inflammation and tissue damage. Viral DNA was only detected in CD206+HLA-DR+CD11b+ cells in ART-treated RMs. The observation of pulmonary and hepatic viral rebound after ART interruption reinforces the importance of early ART implementation to limit viral seeding and inflammatory reactions.
Collapse
Affiliation(s)
- Julien A. Clain
- CHU de Québec Research Center, Laval University, Quebec City, Quebec, Canada
| | | | - Gina Racine
- CHU de Québec Research Center, Laval University, Quebec City, Quebec, Canada
| | - Steven Boutrais
- CHU de Québec Research Center, Laval University, Quebec City, Quebec, Canada
| | | | | | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, University of Quebec in Montreal, Montreal, Quebec, Canada
| | - Arnaud Droit
- CHU de Québec Research Center, Laval University, Quebec City, Quebec, Canada
| | - Petronela Ancuta
- Research Center of the University of Montreal Hospital Center, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology, and Immunology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | | | - Jérôme Estaquier
- CHU de Québec Research Center, Laval University, Quebec City, Quebec, Canada
- INSERM U1124, University of Paris, Paris, France
| |
Collapse
|
9
|
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
|
10
|
Miner MD, Hatherill M, Mave V, Gray GE, Nachman S, Read SW, White RG, Hesseling A, Cobelens F, Patel S, Frick M, Bailey T, Seder R, Flynn J, Rengarajan J, Kaushal D, Hanekom W, Schmidt AC, Scriba TJ, Nemes E, Andersen-Nissen E, Landay A, Dorman SE, Aldrovandi G, Cranmer LM, Day CL, Garcia-Basteiro AL, Fiore-Gartland A, Mogg R, Kublin JG, Gupta A, Churchyard G. Developing tuberculosis vaccines for people with HIV: consensus statements from an international expert panel. Lancet HIV 2022; 9:e791-e800. [PMID: 36240834 PMCID: PMC9667733 DOI: 10.1016/s2352-3018(22)00255-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/16/2022] [Accepted: 08/30/2022] [Indexed: 11/06/2022]
Abstract
New tuberculosis vaccine candidates that are in the development pipeline need to be studied in people with HIV, who are at high risk of acquiring Mycobacterium tuberculosis infection and tuberculosis disease and tend to develop less robust vaccine-induced immune responses. To address the gaps in developing tuberculosis vaccines for people with HIV, a series of symposia was held that posed six framing questions to a panel of international experts: What is the use case or rationale for developing tuberculosis vaccines? What is the landscape of tuberculosis vaccines? Which vaccine candidates should be prioritised? What are the tuberculosis vaccine trial design considerations? What is the role of immunological correlates of protection? What are the gaps in preclinical models for studying tuberculosis vaccines? The international expert panel formulated consensus statements to each of the framing questions, with the intention of informing tuberculosis vaccine development and the prioritisation of clinical trials for inclusion of people with HIV.
Collapse
Affiliation(s)
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Vidya Mave
- Johns Hopkins India, Byramjee-Jeejeebhoy Government Medical College Clinical Research Site, Pune, India
| | - Glenda E Gray
- South African Medical Research Council, Cape Town, South Africa
| | - Sharon Nachman
- Department of Pediatrics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Sarah W Read
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard G White
- Department of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Anneke Hesseling
- Desmond Tutu Tuberculosis Centre, Stellenbosch University, Stellenbosch, South Africa
| | - Frank Cobelens
- Department of Global Health, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sheral Patel
- US Food and Drug Administration, Silver Spring, MD, USA
| | - Mike Frick
- Treatment Action Group, New York, NY, USA
| | | | - Robert Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joanne Flynn
- Microbiology and Molecular Genetics, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Deepak Kaushal
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Willem Hanekom
- Africa Health Research Institute, Durban, KwaZulu-Natal, South Africa
| | | | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Erica Andersen-Nissen
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Cape Town HIV Vaccine Trials Network (HVTN) Immunology Laboratory, Cape Town, South Africa
| | | | - Susan E Dorman
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Grace Aldrovandi
- Department of Pediatrics, University of California, Los Angeles, CA, USA
| | - Lisa M Cranmer
- Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Cheryl L Day
- Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Alberto L Garcia-Basteiro
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Spain; Centro de investigação de Saúde de Manhiça, Maputo, Mozambique
| | | | - Robin Mogg
- Takeda Pharmaceutical Company, Cambridge, MA, USA
| | - James G Kublin
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Amita Gupta
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gavin Churchyard
- The Aurum Institute, Johannesburg, South Africa; School of Public Health, University of Witwatersrand, Johannesburg, South Africa; Department of Medicine, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
11
|
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
|
12
|
Li H, Tang Y, Wang Y, Li Y, Yang Y, Liao K, Song F, Deng S, Chen Y. Single-cell sequencing resolves the landscape of immune cells and regulatory mechanisms in HIV-infected immune non-responders. Cell Death Dis 2022; 13:849. [PMID: 36195585 PMCID: PMC9532384 DOI: 10.1038/s41419-022-05225-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/05/2022]
Abstract
Immune non-responder after highly active antiretroviral therapy (HAART) is the main cause of opportunistic infections and high mortality in AIDS patients, but the mechanism underlying immune reconstitution failure is poorly understood. Here, we performed scRNA-seq, and scATAC-seq analysis of peripheral blood mononuclear cells (PBMCs) derived from immune non-responder (INR) and responder (IR) HIV-1-infected subjects. We found low expression of mucosal-associated invariant T (MAIT) cells in INRs, which exhibited transcriptional profiles associated with impaired mitochondrial function and apoptosis signaling. Single-cell assays for transposase-accessible chromatin (scATAC-seq) and flow cytometry revealed diminished mitochondrial fitness in MAIT cells from INRs, and MAIT had low expression of transcription factor A for mitochondria (TFAM) and peroxisomal proliferator-activated receptor alpha (PPARA). These findings demonstrate that restoring mitochondrial function could modulate the immune dysfunction characteristic of MAIT against bacterial co-infections in INRs subjects.
Collapse
Affiliation(s)
- Haiyu Li
- grid.507893.00000 0004 8495 7810Department of Infectious Disease, Chongqing Public Health Medical Center, 400036 Chongqing, China ,grid.9227.e0000000119573309Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 400714 Chongqing, China
| | - Yongyao Tang
- grid.203458.80000 0000 8653 0555School of Medical Information, Chongqing Medical University, 400016 Chongqing, China
| | - Yujing Wang
- grid.203458.80000 0000 8653 0555School of Medical Information, Chongqing Medical University, 400016 Chongqing, China
| | - Yue Li
- grid.203458.80000 0000 8653 0555Basic Medical College, Chongqing Medical University, 400016 Chongqing, China
| | - Yi Yang
- grid.203458.80000 0000 8653 0555The First Clinical College of Chongqing Medical University, Chongqing Medical University, 400016 Chongqing, China
| | - Kui Liao
- grid.452206.70000 0004 1758 417XDepartment of radiotherapy, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Fangzhou Song
- grid.203458.80000 0000 8653 0555Basic Medical College, Chongqing Medical University, 400016 Chongqing, China
| | - Shixiong Deng
- grid.203458.80000 0000 8653 0555Basic Medical College, Chongqing Medical University, 400016 Chongqing, China
| | - Yaokai Chen
- grid.507893.00000 0004 8495 7810Department of Infectious Disease, Chongqing Public Health Medical Center, 400036 Chongqing, China
| |
Collapse
|
13
|
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
|
14
|
Zheng W, LaCourse SM, Song B, Singh DK, Khanna M, Olivo J, Stern J, Escudero JN, Vergara C, Zhang F, Li S, Wang S, Cranmer LM, Huang Z, Bojanowski CM, Bao D, Njuguna I, Xiao Y, Wamalwa DC, Nguyen DT, Yang L, Maleche-Obimbo E, Nguyen N, Zhang L, Phan H, Fan J, Ning B, Li C, Lyon CJ, Graviss EA, John-Stewart G, Mitchell CD, Ramsay AJ, Kaushal D, Liang R, Pérez-Then E, Hu TY. Diagnosis of paediatric tuberculosis by optically detecting two virulence factors on extracellular vesicles in blood samples. Nat Biomed Eng 2022; 6:979-991. [PMID: 35986185 PMCID: PMC9391224 DOI: 10.1038/s41551-022-00922-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/01/2022] [Indexed: 11/09/2022]
Abstract
Sensitive and specific blood-based assays for the detection of pulmonary and extrapulmonary tuberculosis would reduce mortality associated with missed diagnoses, particularly in children. Here we report a nanoparticle-enhanced immunoassay read by dark-field microscopy that detects two Mycobacterium tuberculosis virulence factors (the glycolipid lipoarabinomannan and its carrier protein) on the surface of circulating extracellular vesicles. In a cohort study of 147 hospitalized and severely immunosuppressed children living with HIV, the assay detected 58 of the 78 (74%) cases of paediatric tuberculosis, 48 of the 66 (73%) cases that were missed by microbiological assays, and 8 out of 10 (80%) cases undiagnosed during the study. It also distinguished tuberculosis from latent-tuberculosis infections in non-human primates. We adapted the assay to make it portable and operable by a smartphone. With further development, the assay may facilitate the detection of tuberculosis at the point of care, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Wenshu Zheng
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sylvia M LaCourse
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Bofan Song
- James C. Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Dhiraj Kumar Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Mayank Khanna
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, LA, USA
| | - Juan Olivo
- O&M Medical School (O&Med), Santo Domingo, Dominican Republic
| | - Joshua Stern
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Jaclyn N Escudero
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Carlos Vergara
- O&M Medical School (O&Med), Santo Domingo, Dominican Republic
| | - Fangfang Zhang
- Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Shaobai Li
- James C. Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Shu Wang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lisa M Cranmer
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Emory School of Medicine, Atlanta, GA, USA
- Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA, USA
| | - Zhen Huang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Christine M Bojanowski
- Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA, USA
| | - Duran Bao
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Irene Njuguna
- Department of Global Health, University of Washington, Seattle, WA, USA
- Kenyatta National Hospital, Research and Programs, Nairobi, Kenya
| | - Yating Xiao
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Dalton C Wamalwa
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Duc T Nguyen
- Department of Pathology and Genomic Medicine, Houston Methodist, Houston, TX, USA
| | - Li Yang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Elizabeth Maleche-Obimbo
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | | | - Lili Zhang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ha Phan
- Center for Promotion of Advancement of Society (CPAS), Ha Noi, Vietnam
- Vietnam National Tuberculosis Program/University of California San Francisco Research Collaboration, Ha Noi, Vietnam
| | - Jia Fan
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bo Ning
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Chenzhong Li
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Christopher J Lyon
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Edward A Graviss
- Department of Pathology and Genomic Medicine, Houston Methodist, Houston, TX, USA
- Department of Surgery, J.C. Walter, Jr. Transplant Center, Sherrie and Alan Conover Center for Liver Disease and Transplantation, Houston Methodist, Houston, TX, USA
| | - Grace John-Stewart
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Charles D Mitchell
- Department of Pediatrics, Division of Infectious Diseases and Immunology, University of Miami Miller School of Medicine, Batchelor Children's Research Institute, Miami, FL, USA
| | - Alistair J Ramsay
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, LA, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Rongguang Liang
- James C. Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Eddy Pérez-Then
- O&M Medical School (O&Med), Santo Domingo, Dominican Republic
| | - Tony Y Hu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA.
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
15
|
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
|
16
|
Gough M, Singh DK, Moodley C, Niu T, Golden NA, Kaushal D, Mehra S. Peripheral Blood Markers Correlate with the Progression of Active Tuberculosis Relative to Latent Control of Mycobacterium tuberculosis Infection in Macaques. Pathogens 2022; 11:544. [PMID: 35631065 PMCID: PMC9146669 DOI: 10.3390/pathogens11050544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 01/08/2023] Open
Abstract
Despite a century of research into tuberculosis (TB), there is a dearth of reproducible, easily quantifiable, biomarkers that can predict disease onset and differentiate between host disease states. Due to the challenges associated with human sampling, nonhuman primates (NHPs) are utilized for recapitulating the closest possible modelling of human TB. To establish a predictive peripheral biomarker profile based on a larger cohort of rhesus macaques (RM), we analyzed results pertaining to peripheral blood serum chemistry and cell counts from RMs that were experimentally exposed to Mtb in our prior studies and characterized as having either developed active TB (ATB) disease or latent TB infection (LTBI). We compared lung CFU burdens and quantitative pathologies with a number of measurables in the peripheral blood. Based on our results, the investigations were then extended to the study of specific molecules and cells in the lung compartments of a subset of these animals and their immune responses. In addition to the elevated serum C-reactive protein (CRP) levels, frequently used to discern the level of Mtb infection in model systems, reduced serum albumin-to-globulin (A/G) ratios were also predictive of active TB disease. Furthermore, higher peripheral myeloid cell levels, particularly those of neutrophils, kynurenine-to-tryptophan ratio, an indicator of induced expression of the immunosuppressive molecule indoleamine dioxygenase, and an influx of myeloid cell populations could also efficiently discriminate between ATB and LTBI in experimentally infected macaques. These quantifiable correlates of disease were then used in conjunction with a regression-based analysis to predict bacterial load. Our results suggest a potential biomarker profile of TB disease in rhesus macaques, that could inform future NHP-TB research. Our results thus suggest that specific biomarkers may be developed from the myeloid subset of peripheral blood or plasma with the ability to discriminate between active and latent Mtb infection.
Collapse
Affiliation(s)
- Maya Gough
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
| | - Dhiraj K. Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
| | - Chivonne Moodley
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
- Tulane University Health Science Center, New Orleans, LA 70112, USA
| | - Tianhua Niu
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Nadia A. Golden
- Tulane National Primate Research Center, Covington, LA 70433, USA;
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (M.G.); (D.K.S.); (C.M.)
| |
Collapse
|
17
|
Liposomal Glutathione Helps to Mitigate Mycobacterium tuberculosis Infection in the Lungs. Antioxidants (Basel) 2022; 11:antiox11040673. [PMID: 35453358 PMCID: PMC9031130 DOI: 10.3390/antiox11040673] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 01/13/2023] Open
Abstract
Mycobacterium tuberculosis (M. tb), the causative agent of tuberculosis (TB), is responsible for causing significant morbidity and mortality, especially among individuals with compromised immune systems. We have previously shown that the supplementation of liposomal glutathione (L-GSH) reduces M. tb viability and enhances a Th-1 cytokine response, promoting granuloma formation in human peripheral blood mononuclear cells in vitro. However, the effects of L-GSH supplementation in modulating the immune responses in the lungs during an active M. tb infection have yet to be explored. In this article, we report the effects of L-GSH supplementation during an active M. tb infection in a mouse model of pulmonary infection. We determine the total GSH levels, malondialdehyde (MDA) levels, cytokine profiles, granuloma formation, and M. tb burden in untreated and L-GSH-treated mice over time. In 40 mM L-GSH-supplemented mice, an increase in the total GSH levels was observed in the lungs. When compared to untreated mice, the treatment of M. tb-infected mice with 40 mM and 80 mM L-GSH resulted in a reduction in MDA levels in the lungs. L-GSH treatment also resulted in a significant increase in the levels of IL-12, IFN-γ, IL-2, IL-17, and TNF-α in the lungs, while down-regulating the production of IL-6, IL-10, and TGF-β in the lungs. A reduction in M. tb survival along with a decrease in granuloma size in the lungs of M. tb-infected mice was observed after L-GSH treatment. Our results show that the supplementation of mice with L-GSH led to increased levels of total GSH, which is associated with reduced oxidative stress, increased levels of granuloma-promoting cytokines, and decreased M. tb burden in the lung. These results illustrate how GSH can help mitigate M. tb infection and provide an insight into future therapeutic interventions.
Collapse
|
18
|
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
|
19
|
Singh DK, Aladyeva E, Das S, Singh B, Esaulova E, Swain A, Ahmed M, Cole J, Moodley C, Mehra S, Schlesinger LS, Artyomov MN, Khader SA, Kaushal D. Myeloid cell interferon responses correlate with clearance of SARS-CoV-2. Nat Commun 2022; 13:679. [PMID: 35115549 PMCID: PMC8814034 DOI: 10.1038/s41467-022-28315-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/04/2022] [Indexed: 01/07/2023] Open
Abstract
Emergence of mutant SARS-CoV-2 strains associated with an increased risk of COVID-19-related death necessitates better understanding of the early viral dynamics, host responses and immunopathology. Single cell RNAseq (scRNAseq) allows for the study of individual cells, uncovering heterogeneous and variable responses to environment, infection and inflammation. While studies have reported immune profiling using scRNAseq in terminal human COVID-19 patients, performing longitudinal immune cell dynamics in humans is challenging. Macaques are a suitable model of SARS-CoV-2 infection. Our longitudinal scRNAseq of bronchoalveolar lavage (BAL) cell suspensions from young rhesus macaques infected with SARS-CoV-2 (n = 6) demonstrates dynamic changes in transcriptional landscape 3 days post- SARS-CoV-2-infection (3dpi; peak viremia), relative to 14-17dpi (recovery phase) and pre-infection (baseline) showing accumulation of distinct populations of both macrophages and T-lymphocytes expressing strong interferon-driven inflammatory gene signature at 3dpi. Type I interferon response is induced in the plasmacytoid dendritic cells with appearance of a distinct HLADR+CD68+CD163+SIGLEC1+ macrophage population exhibiting higher angiotensin-converting enzyme 2 (ACE2) expression. These macrophages are significantly enriched in the lungs of macaques at 3dpi and harbor SARS-CoV-2 while expressing a strong interferon-driven innate anti-viral gene signature. The accumulation of these responses correlated with decline in viremia and recovery.
Collapse
Affiliation(s)
- Dhiraj K Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - Ekaterina Aladyeva
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Shibali Das
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Bindu Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - Ekaterina Esaulova
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Amanda Swain
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Mushtaq Ahmed
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Journey Cole
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - Chivonne Moodley
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - Larry S Schlesinger
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - Maxim N Artyomov
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA.
| |
Collapse
|
20
|
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
|
21
|
Ferreira CM, Barbosa AM, Barreira-Silva P, Silvestre R, Cunha C, Carvalho A, Rodrigues F, Correia-Neves M, Castro AG, Torrado E. Early IL-10 promotes vasculature-associated CD4+ T cells unable to control Mycobacterium tuberculosis infection. JCI Insight 2021; 6:150060. [PMID: 34554927 PMCID: PMC8663558 DOI: 10.1172/jci.insight.150060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/22/2021] [Indexed: 12/29/2022] Open
Abstract
Cytokine-producing CD4+ T cells play a crucial role in the control of Mycobacterium tuberculosis infection; however, there is a delayed appearance of effector T cells in the lungs following aerosol infection. The immunomodulatory cytokine IL-10 antagonizes control of M. tuberculosis infection through mechanisms associated with reduced CD4+ T cell responses. Here, we show that IL-10 overexpression only before the onset of the T cell response impaired control of M. tuberculosis growth; during chronic infection, IL-10 overexpression reduced the CD4+ T cell response without affecting the outcome of infection. IL-10 overexpression early during infection did not, we found, significantly impair the kinetics of CD4+ T cell priming and effector differentiation. However, CD4+ T cells primed and differentiated in an IL-10–enriched environment displayed reduced expression of CXCR3 and, because they did not migrate into the lung parenchyma, their ability to control infection was limited. Importantly, these CD4+ T cells maintained their vasculature phenotype and were unable to control infection, even after adoptive transfer into low IL-10 settings. Together our data support a model wherein, during M. tuberculosis infection, IL-10 acts intrinsically on T cells, impairing their parenchymal migratory capacity and ability to engage with infected phagocytic cells, thereby impeding control of infection.
Collapse
|
22
|
Sharan R, Singh DK, Rengarajan J, Kaushal D. Characterizing Early T Cell Responses in Nonhuman Primate Model of Tuberculosis. Front Immunol 2021; 12:706723. [PMID: 34484203 PMCID: PMC8416058 DOI: 10.3389/fimmu.2021.706723] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/28/2021] [Indexed: 11/21/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a leading infectious disease killer worldwide with 1.4 million TB deaths in 2019. While the majority of infected population maintain an active control of the bacteria, a subset develops active disease leading to mortality. Effective T cell responses are critical to TB immunity with CD4+ and CD8+ T cells being key players of defense. These early cellular responses to TB infection have not yet been studied in-depth in either humans or preclinical animal models. Characterizing early T cell responses in a physiologically relevant preclinical model can provide valuable understanding of the factors that control disease development. We studied Mtb-specific T cell responses in the lung compartment of rhesus macaques infected with either a low- or a high-dose of Mtb CDC1551 via aerosol. Relative to baseline, significantly higher Mtb-specific CD4+IFN-γ+ and TNF-α+ T cell responses were observed in the BAL of low dose infected macaques as early as week 1 post TB infection. The IFN-γ and TNF-a response was delayed to week 3 post infection in Mtb-specific CD4+ and CD8+T cells in the high dose group. The manifestation of earlier T cell responses in the group exposed to the lower Mtb dose suggested a critical role of these cytokines in the antimycobacterial immune cascade, and specifically in the granuloma formation to contain the bacteria. However, a similar increase was not reflected in the CD4+ and CD8+IL-17+ T cells at week 1 post infection in the low dose group. This could be attributed to either a suppression of the IL-17 response or a lack of induction at this early stage of infection. On the contrary, there was a significantly higher IL-17+ response in Mtb-specific CD4+ and CD8+T cells at week 3 in the high dose group. The results clearly demonstrate an early differentiation in the immunity following low dose and high dose infection, largely represented by differences in the IFN-γ and TNF-α response by Mtb-specific T cells in the BAL. This early response to antigen expression by the bacteria could be critical for both bacterial growth control and bacterial containment.
Collapse
Affiliation(s)
- Riti Sharan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Dhiraj Kumar Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jyothi Rengarajan
- Emory Vaccine Center and Yerkes National Primate Research Center (YNPRC), Emory University School of Medicine, Atlanta, GA, United States
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
23
|
Singh D, Aladyeva E, Das S, Singh B, Esaulova E, Swain A, Ahmed M, Cole J, Moodley C, Mehra S, Schlesinger L, Artyomov M, Khader S, Kaushal D. Myeloid cell interferon responses correlate with clearance of SARS-CoV-2. RESEARCH SQUARE 2021:rs.3.rs-664507. [PMID: 34282414 PMCID: PMC8288154 DOI: 10.21203/rs.3.rs-664507/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The emergence of mutant SARS-CoV-2 strains associated with an increased risk of COVID-19-related death necessitates better understanding of the early viral dynamics, host responses and immunopathology. While studies have reported immune profiling using single cell RNA sequencing in terminal human COVID-19 patients, performing longitudinal immune cell dynamics in humans is challenging. Macaques are a suitable model of SARS-CoV-2 infection. We performed longitudinal single-cell RNA sequencing of bronchoalveolar lavage (BAL) cell suspensions from adult rhesus macaques infected with SARS-CoV-2 (n=6) to delineate the early dynamics of immune cells changes. The bronchoalveolar compartment exhibited dynamic changes in transcriptional landscape 3 days post- SARS-CoV-2-infection (3dpi) (peak viremia), relative to 14-17dpi (recovery phase) and pre-infection (baseline). We observed the accumulation of distinct populations of both macrophages and T-lymphocytes expressing strong interferon-driven inflammatory gene signature at 3dpi. Type I IFN response was highly induced in the plasmacytoid dendritic cells. The presence of a distinct HLADR+CD68+CD163+SIGLEC1+ macrophage population exhibiting higher angiotensin converting enzyme 2 (ACE2) expression was also observed. These macrophages were significantly recruited to the lungs of macaques at 3dpi and harbored SARS-CoV-2, while expressing a strong interferon-driven innate anti-viral gene signature. The accumulation of these responses correlated with decline in viremia and recovery. The recruitment of a myeloid cell-mediated Type I IFN response is associated with the rapid clearance of SARS-CoV-2 infection in macaques.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Larry Schlesinger
- Southwest National Primate Research Center Texas Biomedical Research Institute
| | | | | | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute
| |
Collapse
|
24
|
Zhou MJ, Huang HH, Song JW, Tu B, Fan X, Li J, Jin JH, Cao WJ, Hu W, Yang T, Zhou CB, Yuan JH, Fan J, Zhang JY, Jiao YM, Xu RN, Zhen C, Shi M, Zhang C, Wang FS. Compromised long-lived memory CD8 + T cells are associated with reduced IL-7 responsiveness in HIV-infected immunological nonresponders. Eur J Immunol 2021; 51:2027-2039. [PMID: 33974710 DOI: 10.1002/eji.202149203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/06/2021] [Indexed: 11/07/2022]
Abstract
Immune deficiency is one of the hallmarks of HIV infection and a major cause of adverse outcomes in people living with HIV (PLWH). Long-lived memory CD8+ T cells (LLMCs) are essential executors of long-term protective immunity; however, the generation and maintenance of LLMCs during chronic HIV infection are not well understood. In the present study, we analyzed circulating LLMCs in healthy controls (HCs) and PLWH with different disease statuses, including treatment naïve patients (TNs), complete responders (CRs), and immunological nonresponders (INRs). We found that both TNs and INRs showed severely compromised LLMCs compared with HCs and CRs, respectively. The decrease of LLMCs in TNs correlated positively with the reduction of their precursors, namely memory precursor effector T cells (MPECs), which might be associated with elevated pro-inflammatory cytokines. Strikingly, INRs showed an accumulation of MPECs, which exhibited diminished responsiveness to interleukin 7 (IL-7), thereby indicating abrogated differentiation into LLMCs. Moreover, in vitro studies showed that treatment with dexamethasone could improve the IL7-phosphorylated (p)-signal transducer and activator of transcription (STAT5) response by upregulating the expression of the interleukin 7 receptor (IL-7Rα) on MPECs in INRs. These findings provide insights that will encourage the development of novel therapeutics to improve immune function in PLWH.
Collapse
Affiliation(s)
- Ming-Ju Zhou
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Hui-Huang Huang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin-Wen Song
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Bo Tu
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xing Fan
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jing Li
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- 302 Clinical Medical School, Peking University, Beijing, China
| | - Jie-Hua Jin
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Wen-Jing Cao
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Bengbu Medical College, Bengbu, China
| | - Wei Hu
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Tao Yang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Chun-Bao Zhou
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin-Hong Yuan
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin Fan
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ji-Yuan Zhang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yan-Mei Jiao
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ruo-Nan Xu
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Cheng Zhen
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ming Shi
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chao Zhang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Fu-Sheng Wang
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- 302 Clinical Medical School, Peking University, Beijing, China
- Bengbu Medical College, Bengbu, China
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
25
|
Van de Wijer L, van der Heijden WA, Ter Horst R, Jaeger M, Trypsteen W, Rutsaert S, van Cranenbroek B, van Rijssen E, Joosten I, Joosten L, Vandekerckhove L, Schoofs T, van Lunzen J, Netea MG, Koenen HJPM, van der Ven AJAM, de Mast Q. The Architecture of Circulating Immune Cells Is Dysregulated in People Living With HIV on Long Term Antiretroviral Treatment and Relates With Markers of the HIV-1 Reservoir, Cytomegalovirus, and Microbial Translocation. Front Immunol 2021; 12:661990. [PMID: 33953724 PMCID: PMC8091964 DOI: 10.3389/fimmu.2021.661990] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/29/2021] [Indexed: 12/26/2022] Open
Abstract
Long-term changes in the immune system of successfully treated people living with HIV (PLHIV) remain incompletely understood. In this study, we assessed 108 white blood cell (WBC) populations in a cohort of 211 PLHIV on stable antiretroviral therapy and in 56 HIV-uninfected controls using flow cytometry. We show that marked differences exist in T cell maturation and differentiation between PLHIV and HIV-uninfected controls: PLHIV had reduced percentages of CD4+ T cells and naïve T cells and increased percentages of CD8+ T cells, effector T cells, and T helper 17 (Th17) cells, together with increased Th17/regulatory T cell (Treg) ratios. PLHIV also exhibited altered B cell maturation with reduced percentages of memory B cells and increased numbers of plasmablasts. Determinants of the T and B cell composition in PLHIV included host factors (age, sex, and smoking), markers of the HIV reservoir, and CMV serostatus. Moreover, higher circulating Th17 percentages were associated with higher plasma concentrations of interleukin (IL) 6, soluble CD14, the gut homing chemokine CCL20, and intestinal fatty acid binding protein (IFABP). The changes in circulating lymphocytes translated into functional changes with reduced interferon (IFN)- γ responses of peripheral blood mononuclear cells to stimulation with Candida albicans and Mycobacterium tuberculosis. In conclusion, this comprehensive analysis confirms the importance of persistent abnormalities in the number and function of circulating immune cells in PLHIV on stable treatment.
Collapse
Affiliation(s)
- Lisa Van de Wijer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Wouter A van der Heijden
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rob Ter Horst
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Martin Jaeger
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Wim Trypsteen
- HIV Cure Research Center, Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Sofie Rutsaert
- HIV Cure Research Center, Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Bram van Cranenbroek
- Laboratory for Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Esther van Rijssen
- Laboratory for Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Irma Joosten
- Laboratory for Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Leo Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, Ghent, Belgium
| | | | | | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences 12 Institute (LIMES), University of Bonn, Bonn, Germany
| | - Hans J P M Koenen
- Laboratory for Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - André J A M van der Ven
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
26
|
Sterling TR, Lin PL. Treatment of latent M. tuberculosis infection and use of antiretroviral therapy to prevent tuberculosis. J Clin Invest 2021; 130:5102-5104. [PMID: 32831291 DOI: 10.1172/jci137294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Even with treatment of HIV with antiretroviral therapy (ART), the risk of tuberculosis (TB) reactivation remains higher in HIV-infected than HIV-uninfected persons. In this issue of the JCI, Ganatra et al. explored TB reactivation in the context of ART using TB and simian immunodeficiency virus-coinfected (TB/SIV-coinfected) nonhuman primates. The authors found that treating rhesus macaques with ART restored CD4+ T cells in whole blood, spleen, and bronchoalveolar lavage (BAL) fluid, but not in the lung interstitium. TB risk was not decreased in the coinfected macaques treated with ART for 14-63 days, suggesting that ART does not decrease the short-term risk of reactivation. Reactivation occurred as CD4+ T cells were increasing, which is consistent with observations made in humans. This study provides a potential model for systematic evaluation of TB/SIV coinfection and different treatment regimens and strategies to prevent TB reactivation.
Collapse
Affiliation(s)
- Timothy R Sterling
- Vanderbilt University Medical Center, Division of Infectious Diseases, Department of Medicine, Nashville, Tennessee, USA
| | - Philana Ling Lin
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
27
|
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
|
28
|
Chetty A, Omondi MA, Butters C, Smith KA, Katawa G, Ritter M, Layland L, Horsnell W. Impact of Helminth Infections on Female Reproductive Health and Associated Diseases. Front Immunol 2020; 11:577516. [PMID: 33329545 PMCID: PMC7719634 DOI: 10.3389/fimmu.2020.577516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/27/2020] [Indexed: 12/25/2022] Open
Abstract
A growing body of knowledge exists on the influence of helminth infections on allergies and unrelated infections in the lung and gastrointestinal (GI) mucosa. However, the bystander effects of helminth infections on the female genital mucosa and reproductive health is understudied but important considering the high prevalence of helminth exposure and sexually transmitted infections in low- and middle-income countries (LMICs). In this review, we explore current knowledge about the direct and systemic effects of helminth infections on unrelated diseases. We summarize host disease-controlling immunity of important sexually transmitted infections and introduce the limited knowledge of how helminths infections directly cause pathology to female reproductive tract (FRT), alter susceptibility to sexually transmitted infections and reproduction. We also review work by others on type 2 immunity in the FRT and hypothesize how these insights may guide future work to help understand how helminths alter FRT health.
Collapse
Affiliation(s)
- Alisha Chetty
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Millicent A Omondi
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Claire Butters
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Katherine Ann Smith
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Gnatoulma Katawa
- Ecole Supérieure des Techniques Biologiques et Alimentaires, Université de Lomé, Lomé, Togo
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - Laura Layland
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - William Horsnell
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
29
|
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
|