1
|
Yanagihara T, Hata K, Matsubara K, Kunimura K, Suzuki K, Tsubouchi K, Ikegame S, Fukui Y, Okamoto I. Immunophenotyping of T Cells in Lung Malignancies and Cryptogenic Organizing Pneumonia. J Clin Med 2025; 14:316. [PMID: 39860323 PMCID: PMC11766438 DOI: 10.3390/jcm14020316] [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: 11/28/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Lung malignancies, including cancerous lymphangitis and lymphomas, can mimic interstitial lung diseases like cryptogenic organizing pneumonia (COP) on imaging, leading to diagnostic delays. We aimed to identify potential biomarkers to distinguish between these conditions. Methods: We analyzed bronchoalveolar lavage fluid from 8 patients (4 COP, mean age 59.8 ± 13.5 years; 4 lung malignancies including 2 cancerous lymphangitis, 1 MALT lymphoma, and 1 diffuse large B cell lymphoma, mean age 67.8 ± 4.5 years) using mass cytometry with 35 T cell markers. Data were analyzed using principal component analysis (PCA) and unsupervised Citrus clustering. Results: PCA of T cell marker intensities effectively separated the two groups, with IL-2Rα, PD-L2, CD45RA, CD44, and OX40 being the top discriminating markers. Citrus analysis showed a significant increase in the CD16+ CD4+ and CD16+ CD8+ T cell populations in the COP group compared to lung malignancies. Conclusions: Our findings reveal distinct T cell immunophenotypes in COP versus lung malignancies, particularly increased CD16+ T cells in COP, which could serve as potential diagnostic biomarkers.
Collapse
Affiliation(s)
- Toyoshi Yanagihara
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Department of Respiratory Medicine, Fukuoka University Hospital, Fukuoka 814-0180, Japan
| | - Kentaro Hata
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Keisuke Matsubara
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kazufumi Kunimura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kunihiro Suzuki
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kazuya Tsubouchi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Satoshi Ikegame
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
2
|
Ssekamatte P, Nabatanzi R, Sitenda D, Nakibuule M, Bagaya BS, Kibirige D, Kyazze AP, Kateete DP, Sande OJ, van Crevel R, Cose S, Biraro IA. Impaired Mycobacterium tuberculosis-specific T-cell memory phenotypes and functional profiles among adults with type 2 diabetes mellitus in Uganda. Front Immunol 2024; 15:1480739. [PMID: 39430752 PMCID: PMC11486641 DOI: 10.3389/fimmu.2024.1480739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Background Efforts to eradicate tuberculosis (TB) are threatened by diabetes mellitus (DM), which confers a 3-fold increase in the risk of TB disease. The changes in the memory phenotypes and functional profiles of Mycobacterium tuberculosis (Mtb)-specific T cells in latent TB infection (LTBI)-DM participants remain poorly characterised. We, therefore, assessed the effect of DM on T-cell phenotype and function in LTBI and DM clinical groups. Methods We compared the memory phenotypes and function profiles of Mtb-specific CD4+ and CD8+ T cells among participants with LTBI-DM (n=21), LTBI-only (n=17) and DM-only (n=16). Peripheral blood mononuclear cells (PBMCs) were stimulated with early secretory antigenic 6 kDa (ESAT-6) and culture filtrate protein 10 (CFP-10) peptide pools or phytohemagglutinin (PHA). The memory phenotypes (CCR7/CD45RA), and functional profiles (HLA-DR, PD-1, CD107a, IFN-γ, IL-2, TNF, IL-13, IL-17A) of Mtb-specific CD4+ and CD8+ T cells were characterised by flow cytometry. Results Naïve CD4+ T cells were significantly decreased in the LTBI-DM compared to the LTBI-only participants [0.47 (0.34-0.69) vs 0.91 (0.59-1.05); (p<0.001)]. Similarly, CD8+ HLA-DR expression was significantly decreased in LTBI-DM compared to LTBI-only participants [0.26 (0.19-0.33) vs 0.52 (0.40-0.64); (p<0.0001)], whereas CD4+ and CD8+ PD-1 expression was significantly upregulated in the LTBI-DM compared to the LTBI-only participants [0.61 (0.53-0.77) vs 0.19 (0.10-0.28); (p<0.0001) and 0.41 (0.37-0.56) vs 0.29 (0.17-0.42); (p=0.007)] respectively. CD4+ and CD8+ IFN-γ production was significantly decreased in the LTBI-DM compared to the LTBI-only participants [0.28 (0.19-0.38) vs 0.39 (0.25-0.53); (p=0.030) and 0.36 (0.27-0.49) vs 0.55 (0.41-0.88); (p=0.016)] respectively. CD4+ TNF and CD8+ IL-17A production were significantly decreased in participants with LTBI-DM compared to those with LTBI-only [0.38 (0.33-0.50) vs 0.62 (0.46-0.87); (p=0.004) and 0.29 (0.16-0.42) vs 0.47 (0.29-0.52); (0.017)] respectively. LTBI-DM participants had significantly lower dual-functional (IFN-γ+IL-2+ and IL-2+TNF+) and mono-functional (IFN-γ+ and TNF+) CD4+ responses than LTBI-only participants. LTBI-DM participants had significantly decreased dual-functional (IFN-γ+IL-2+, IFN-γ+ TNF+ and IL-2+TNF+) and mono-functional (IFN-γ+, IL-2+ and TNF+) central and effector memory CD4+ responses compared to LTBI-only participants. Conclusion Type 2 DM impairs the memory phenotypes and functional profiles of Mtb-specific CD4+ and CD8+ T cells, potentially indicating underlying immunopathology towards increased active TB disease risk.
Collapse
Affiliation(s)
- Phillip Ssekamatte
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Entebbe, Uganda
| | - Rose Nabatanzi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Diana Sitenda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Marjorie Nakibuule
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Entebbe, Uganda
| | - Bernard Ssentalo Bagaya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Davis Kibirige
- Department of Medicine, Uganda Martyrs Lubaga Hospital, Kampala, Uganda
| | - Andrew Peter Kyazze
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - David Patrick Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Obondo James Sande
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Stephen Cose
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Entebbe, Uganda
| | - Irene Andia Biraro
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Entebbe, Uganda
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
3
|
Lan Z, Wei Y, Yue K, He R, Jiang Z. Genetically predicted immune cells mediate the association between gut microbiota and neuropathy pain. Inflammopharmacology 2024; 32:3357-3373. [PMID: 38955934 PMCID: PMC11416384 DOI: 10.1007/s10787-024-01514-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Previous observational studies have indicated a complex association between gut microbiota (GM) and neuropathic pain (NP). Nonetheless, the precise biological mechanisms underlying this association remain unclear. Therefore, we adopted a Mendelian randomization (MR) approach to investigate the causal relationship between GM and neuropathic pain including post-herpetic neuralgia (PHN), painful diabetic peripheral neuropathy (PDPN), and trigeminal neuralgia (TN), as well as to explore the potential mediation effects of immune cells. METHODS We performed a two-step, two-sample Mendelian randomization study with an inverse variance-weighted (IVW) approach to investigate the causal role of GM on three major kinds of NP and the mediation effect of immune cells between the association of GM and NP. In addition, we determine the strongest causal associations using Bayesian weighted Mendelian randomization (BWMR) analysis. Furthermore, we will investigate the mediating role of immune cells through a two-step Mendelian randomization design. RESULTS We identified 53 taxonomies and pathways of gut microbiota that had significant causal associations with NP. In addition, we also discovered 120 immune cells that exhibited significant causal associations with NP. According to the BWMR and two-step Mendelian randomization analysis, we identified the following results CD4 on CM CD4 + (maturation stages of T cell) mediated 6.7% of the risk reduction for PHN through the pathway of fucose degradation (FUCCAT.PWY). CD28 + DN (CD4-CD8-) AC (Treg) mediated 12.5% of the risk reduction for PHN through the influence on Roseburia inulinivorans. CD45 on lymphocyte (Myeloid cell) mediated 11.9% of the risk increase for TN through the superpathway of acetyl-CoA biosynthesis (PWY.5173). HLA DR + CD8br %T cell (TBNK) mediated 3.2% of the risk reduction for TN through the superpathway of GDP-mannose-derived O-antigen building blocks biosynthesis (PWY.7323). IgD-CD38-AC (B cell) mediated 7.5% of the risk reduction for DPN through the pathway of thiazole biosynthesis I in E. coli (PWY.6892). DISCUSSION These findings provided evidence supporting the causal effect of GM with NP, with immune cells playing a mediating role. These findings may inform prevention strategies and interventions directed toward NP. Future studies should explore other plausible biological mechanisms.
Collapse
Affiliation(s)
- Zhixuan Lan
- Department of Pain Medicine, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, Nanning, 530005, China
| | - Yi Wei
- Department of Pain Medicine, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, Nanning, 530005, China
| | - Kan Yue
- Department of Pain Medicine, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, Nanning, 530005, China
| | - Ruilin He
- Department of Pain Medicine, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, Nanning, 530005, China.
| | - Zongbin Jiang
- Department of Pain Medicine, The Second Affiliated Hospital of Guangxi Medical University, Guangxi, Nanning, 530005, China.
| |
Collapse
|
4
|
Yang Y, Shi H, Zhou Y, Zhou Y. Expression of HLA-DR and KLRG1 enhances the cytotoxic potential and cytokine secretion capacity of CD3 + T cells in tuberculosis patients. Int Immunopharmacol 2024; 133:112115. [PMID: 38652959 DOI: 10.1016/j.intimp.2024.112115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/26/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Human T cells play an important role in immunity against tuberculosis (TB) infection. Activating receptor HLA-DR and inhibitory receptor KLRG1 are critical regulators of T cell function during viral infection and tumorigenesis, but they have been less studied in TB infection. METHODS In this study, we explored the relationship between CD3+ T cell expression of HLA-DR and KLRG1 receptors and function against TB infection. Flow cytometry was conducted to assess the immunomodulatory effects of HLA-DR and KLRG1 receptors on CD3+ T cells in patients with different TB infection status. RESULTS We found activating receptors HLA-DR, NKG2C, CD57 and NKP46, and inhibitory receptors KLRG1 and KIR on CD3+ T cells in different TB infection status showed different distribution patterns; the cytotoxic potential and cytokine secretion capacity of CD3+ T cells after Mtb-specific antigen stimulation were significantly enhanced in TB infection groups. Further studies revealed HLA-DR+ T and KLRG1+ T cells expressed higher activating and inhibitory receptors than the negative population. In addition, the expression of cytotoxic potential and cytokine secretion capacity of HLA-DR+ T and KLRG1+ T cells was significantly higher than that of HLA-DR- T and KLRG1- T cells. CONCLUSIONS Expression of HLA-DR and KLRG1 enhances the cytotoxic potential and cytokine secretion capacity of CD3+ T cells in TB patients, suggesting CD3+ T cells expressing HLA-DR and KLRG1 are important effector cell phenotypes involved in the host anti-TB infection. HLA-DR and KLRG1 expressed by CD3+ T cells may be potential predictive markers of TB disease progression and clinical immune assessment.
Collapse
Affiliation(s)
- Yiqi Yang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
| | - Hanlu Shi
- Clinical Research Center, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 360000, China
| | - Yu Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| | - Yonglie Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
5
|
Gootjes C, Zwaginga JJ, Roep BO, Nikolic T. Defining Human Regulatory T Cells beyond FOXP3: The Need to Combine Phenotype with Function. Cells 2024; 13:941. [PMID: 38891073 PMCID: PMC11172350 DOI: 10.3390/cells13110941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/18/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Regulatory T cells (Tregs) are essential to maintain immune homeostasis by promoting self-tolerance. Reduced Treg numbers or functionality can lead to a loss of tolerance, increasing the risk of developing autoimmune diseases. An overwhelming variety of human Tregs has been described, based on either specific phenotype, tissue compartment, or pathological condition, yet the bulk of the literature only addresses CD25-positive and CD127-negative cells, coined by naturally occurring Tregs (nTregs), most of which express the transcription factor Forkhead box protein 3 (FOXP3). While the discovery of FOXP3 was seminal to understanding the origin and biology of nTregs, there is evidence in humans that not all T cells expressing FOXP3 are regulatory, and that not all Tregs express FOXP3. Namely, the activation of human T cells induces the transient expression of FOXP3, irrespective of whether they are regulatory or inflammatory effectors, while some induced T cells that may be broadly defined as Tregs (e.g., Tr1 cells) typically lack demethylation and do not express FOXP3. Furthermore, it is unknown whether and how many nTregs exist without FOXP3 expression. Several other candidate regulatory molecules, such as GITR, Lag-3, GARP, GPA33, Helios, and Neuropilin, have been identified but subsequently discarded as Treg-specific markers. Multiparametric analyses have uncovered a plethora of Treg phenotypes, and neither single markers nor combinations thereof can define all and only Tregs. To date, only the functional capacity to inhibit immune responses defines a Treg and distinguishes Tregs from inflammatory T cells (Teffs) in humans. This review revisits current knowledge of the Treg universe with respect to their heterogeneity in phenotype and function. We propose that it is unavoidable to characterize human Tregs by their phenotype in combination with their function, since phenotype alone does not unambiguously define Tregs. There is an unmet need to align the expression of specific markers or combinations thereof with a particular suppressive function to coin functional Treg entities and categorize Treg diversity.
Collapse
Affiliation(s)
- Chelsea Gootjes
- Laboratory of Immunomodulation and Regenerative Cell Therapy, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (J.J.Z.); (T.N.)
| | | | | | | |
Collapse
|
6
|
Setlhare B, Letsoalo M, Nkabinde SA, Nkabinde M, Mzobe G, Mtshali A, Parveen S, Ngcobo S, Invernizzi L, Maharaj V, Ngcobo M, Gqaleni N. An in vitro study to elucidate the effects of product Nkabinde on immune response in peripheral blood mononuclear cells of healthy donors. Front Pharmacol 2024; 15:1308913. [PMID: 38533263 PMCID: PMC10963514 DOI: 10.3389/fphar.2024.1308913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Introduction: A significant number of the South African population still rely on traditional medicines (TM) as their primary healthcare due to their belief in their holistic healing and immune-boosting properties. However, little to no scientific data is available on the effects of most TM products on cytokine and cellular biomarkers of the immune response. Here, we evaluated the impact of traditional medicine [Product Nkabinde (PN)] in inducing cellular and cytokine biomarkers of inflammation in peripheral blood mononuclear cells (PBMCs) from eight healthy volunteers. Methods: PN was supplied by a local Traditional Health Practitioner (THP). The IC50 (half maximum concentration) of the standardized extract on isolated PBMCs was established using the cell viability assay over 24 h of incubation. Luminex and flow cytometry assays were used to measure cytokine and cellular levels in PBMCs stimulated with PN and/or PHA over 24, 48, and 72 h, respectively. Results: The IC50 concentration of PN in treated PBMCs was established at 325.3 μg/mL. In the cellular activation assay, the percentages of CD38-HLA-DR + on total CD4+ T cells were significantly increased in PBMCs stimulated with PN compared to unstimulated controls after 24 h (p = 0.008). PN significantly induced the production of anti-inflammatory IL-10 (p = 0.041); proinflammatory cytokines IL-1α (p = 0.003), TNF-α (p < 0.0001); and chemokine MIP-1β (p = 0.046) compared to the unstimulated control after 24 h. At 48 h incubation, the production of proinflammatory cytokines IL-1α (p = 0.034) and TNF-α (p = 0.011) were significantly induced following treatment with PN. Conclusion: We conclude that the PN possesses in vitro immunomodulatory properties that may influence immune and inflammatory responses. More studies using PN are needed to further understand key parameters mediating induction, expression, and regulation of the immune response in the context of pathogen-associated infections.
Collapse
Affiliation(s)
- Boitumelo Setlhare
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Marothi Letsoalo
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Siphathimandla Authority Nkabinde
- Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Magugu Nkabinde
- Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Gugulethu Mzobe
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Mtshali
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Sobia Parveen
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Samukelisiwe Ngcobo
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Centre for Aids Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Luke Invernizzi
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Vinesh Maharaj
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Mlungisi Ngcobo
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Nceba Gqaleni
- Discipline of Traditional Medicine, University of KwaZulu-Natal, Durban, South Africa
- Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
7
|
Ahmed A, Tripathi H, van Meijgaarden KE, Kumar NC, Adiga V, Rakshit S, Parthiban C, Eveline J S, D’Souza G, Dias M, Ottenhoff TH, Netea MG, Joosten SA, Vyakarnam A. BCG revaccination in adults enhances pro-inflammatory markers of trained immunity along with anti-inflammatory pathways. iScience 2023; 26:107889. [PMID: 37817935 PMCID: PMC10561055 DOI: 10.1016/j.isci.2023.107889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/22/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023] Open
Abstract
This study characterized mechanisms of Bacille Calmette-Guérin (BCG) revaccination-induced trained immunity (TI) in India. Adults, BCG vaccinated at birth, were sampled longitudinally before and after a second BCG dose. BCG revaccination significantly elevated tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, and IL-6 in HLA-DR+CD16-CD14hi monocytes, demonstrating induction of TI. Mycobacteria-specific CD4+ T cell interferon (IFN) γ, IL-2, and TNF-α were significantly higher in re-vaccinees and correlated positively with HLA-DR+CD16-CD14hi TI responses. This, however, did not translate into increased mycobacterial growth control, measured by mycobacterial growth inhibition assay (MGIA). Post revaccination, elevated secreted TNF-α, IL-1β, and IL-6 to "heterologous" fungal, bacterial, and enhanced CXCL-10 and IFNα to viral stimuli were also observed concomitant with increased anti-inflammatory cytokine, IL-1RA. RNA sequencing after revaccination highlighted a BCG and LPS induced signature which included upregulated IL17 and TNF pathway genes and downregulated key inflammatory genes: CXCL11, CCL24, HLADRA, CTSS, CTSC. Our data highlight a balanced immune response comprising pro- and anti-inflammatory mediators to be a feature of BCG revaccination-induced immunity.
Collapse
Affiliation(s)
- Asma Ahmed
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - Himanshu Tripathi
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | | | - Nirutha Chetan Kumar
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - Vasista Adiga
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
- Department of Biotechnology, PES University, Bangalore, India
| | - Srabanti Rakshit
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - Chaitra Parthiban
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - Sharon Eveline J
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - George D’Souza
- Department of Pulmonary Medicine, St. John’s Medical College, Bangalore, India
| | - Mary Dias
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
| | - Tom H.M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Annapurna Vyakarnam
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Infectious Disease Unit, St. John’s Research Institute, Bangalore, India
- Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Science & Medicine, King’s College, London, UK
| |
Collapse
|
8
|
Starshinova A, Kudryavtsev I, Rubinstein A, Malkova A, Dovgaluk I, Kudlay D. Tuberculosis and COVID-19 Dually Affect Human Th17 Cell Immune Response. Biomedicines 2023; 11:2123. [PMID: 37626620 PMCID: PMC10452633 DOI: 10.3390/biomedicines11082123] [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: 05/31/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19 infection not only profoundly impacts the detection of tuberculosis infection (Tbc) but also affects modality in tuberculosis patient immune response. It is important to determine immune response alterations in latent tuberculosis infection as well as in SARS-CoV-2-infected tuberculosis patients. Such changes may have underlying effects on the development and course of further tuberculosis. Here, we aimed to review the characteristics of immune response in TB patients or convalescent COVID-19 patients with latent TB infection (LTBI). MATERIALS AND METHODS We analyzed the features of immune response in tuberculosis and COVID-19 patients. For this, we analyzed publications released from December 2019 to March 2023; those which were published in accessible international databases ("Medline", "PubMed", "Scopus") and with keywords such as "COVID-19", "SARS-CoV-2", "tuberculosis", "pulmonary tuberculosis", "latent tuberculosis infection", "Treg", "follicular Treg", and "Treg subsets", we considered. RESULTS Through our analysis, we found that tuberculosis patients who had been infected with COVID-19 previously and elevated Th1 and Th2 cell levels. High levels of Th1 and Th2 cells may serve as a positive marker, characterizing activated immune response during TB infection. COVID-19 or post-COVID-19 subjects showed decreased Th17 levels, indicating a lack of tuberculosis development. Moreover, the typical course of tuberculosis is associated with an increase in Treg level, but COVID-19 contributes to a hyperinflammatory response. CONCLUSION According to the data obtained, the course of tuberculosis proceeds in a dissimilar way due to the distinct immune response, elicited by SARS-CoV-2. Importantly, the development of active tuberculosis with a severe course is associated with a decline in Treg levels. Both pathogens lead to disturbed immune responses, increasing the risk of developing severe TB. The insights and findings of this paper may be used to improve the future management of individuals with latent and active tuberculosis.
Collapse
Affiliation(s)
- Anna Starshinova
- Almazov National Medical Research Centre, 197341 St-Petersburg, Russia
| | - Igor Kudryavtsev
- Department of Immunology, Institution of Experimental Medicine, 197022 St-Petersburg, Russia; (I.K.); (A.R.)
| | - Artem Rubinstein
- Department of Immunology, Institution of Experimental Medicine, 197022 St-Petersburg, Russia; (I.K.); (A.R.)
| | - Anna Malkova
- Faculty of Natural Sciences, Ariel University, Ariel 40700, Israel;
| | - Irina Dovgaluk
- Phthisiopulmonology Department, Research Institute of Phthisiopulmonology, 191036 St-Petersburg, Russia;
| | - Dmitry Kudlay
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia;
- Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
| |
Collapse
|
9
|
Mbambo G, Dwivedi A, Ifeonu OO, Munro JB, Shrestha B, Bromley RE, Hodges T, Adkins RS, Kouriba B, Diarra I, Niangaly A, Kone AK, Coulibaly D, Traore K, Dolo A, Thera MA, Laurens MB, Doumbo OK, Plowe CV, Berry AA, Travassos M, Lyke KE, Silva JC. Immunogenomic profile at baseline predicts host susceptibility to clinical malaria. Front Immunol 2023; 14:1179314. [PMID: 37465667 PMCID: PMC10351378 DOI: 10.3389/fimmu.2023.1179314] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/19/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction Host gene and protein expression impact susceptibility to clinical malaria, but the balance of immune cell populations, cytokines and genes that contributes to protection, remains incompletely understood. Little is known about the determinants of host susceptibility to clinical malaria at a time when acquired immunity is developing. Methods We analyzed peripheral blood mononuclear cells (PBMCs) collected from children who differed in susceptibility to clinical malaria, all from a small town in Mali. PBMCs were collected from children aged 4-6 years at the start, peak and end of the malaria season. We characterized the immune cell composition and cytokine secretion for a subset of 20 children per timepoint (10 children with no symptomatic malaria age-matched to 10 children with >2 symptomatic malarial illnesses), and gene expression patterns for six children (three per cohort) per timepoint. Results We observed differences between the two groups of children in the expression of genes related to cell death and inflammation; in particular, inflammatory genes such as CXCL10 and STAT1 and apoptotic genes such as XAF1 were upregulated in susceptible children before the transmission season began. We also noted higher frequency of HLA-DR+ CD4 T cells in protected children during the peak of the malaria season and comparable levels cytokine secretion after stimulation with malaria schizonts across all three time points. Conclusion This study highlights the importance of baseline immune signatures in determining disease outcome. Our data suggests that differences in apoptotic and inflammatory gene expression patterns can serve as predictive markers of susceptibility to clinical malaria.
Collapse
Affiliation(s)
- Gillian Mbambo
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ankit Dwivedi
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Olukemi O. Ifeonu
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - James B. Munro
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Robin E. Bromley
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Theresa Hodges
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ricky S. Adkins
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bourema Kouriba
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Issa Diarra
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Abdoulaye K. Kone
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Drissa Coulibaly
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Karim Traore
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Amagana Dolo
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Mahamadou A. Thera
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ogobara K. Doumbo
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Christopher V. Plowe
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Andrea A. Berry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mark Travassos
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Global Health and Tropical Medicine, Instituto deHigiene e Medicina Tropical, Universidade Nova de Lisboa (GHTM, IHMT, UNL), Lisboa, Portugal
| |
Collapse
|
10
|
The Regulatory-T-Cell Memory Phenotype: What We Know. Cells 2022; 11:cells11101687. [PMID: 35626725 PMCID: PMC9139615 DOI: 10.3390/cells11101687] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 01/25/2023] Open
Abstract
In immunology, the discovery of regulatory T (Treg) cells was a major breakthrough. Treg cells play a key role in pregnancy maintenance, in the prevention of autoimmune responses, and in the control of all immune responses, including responses to self cells, cancer, infection, and a transplant. It is currently unclear whether Treg cells are capable of long-term memory of an encounter with an antigen. Although the term “immunological memory” usually means an enhanced ability to protect the body from reinfection, the memory of the suppressive activity of Treg cells helps to avoid the state of generalized immunosuppression that may result from the second activation of the immune system. In this review, we would like to discuss the concept of regulatory memory and in which tissues memory Treg cells can perform their functions.
Collapse
|
11
|
Polak D, Bohle B. Neutrophils-typical atypical antigen presenting cells? Immunol Lett 2022; 247:52-58. [DOI: 10.1016/j.imlet.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 11/05/2022]
|
12
|
Frequency of CD4+ regulatory T cells and modulation of CD4+T lymphocyte activation in pleural tuberculoma. Tuberculosis (Edinb) 2022; 134:102210. [DOI: 10.1016/j.tube.2022.102210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/17/2022] [Accepted: 04/24/2022] [Indexed: 11/21/2022]
|
13
|
A Novel Approach for Fast Screening of a Complex Cyanobacterial Extract for Immunomodulatory Properties and Antibacterial Activity. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12062847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The filamentous cyanobacteria from genus Phormidium are rich natural sources of bioactive compounds that could be exploited as pharmaceuticals or nutraceuticals. In this study, we suggest a novel approach for assessing the immunomodulatory properties of the products derived from cyanobacteria. The influence of Phormidium papyraceum extract on the human leukocyte immunophenotype was evaluated by attempting to link this activity to certain putative compounds identified in the extract. By using three staining panels and flow cytometry, we found that the cyanobacterial extract affected mainly CD4+ T cells upregulating activated CD4+CD152+ T cells (15.75 ± 1.93% treated vs. 4.65 ± 1.41% control) and regulatory CD4+CD25+ T cells (5.36 ± 0.64% treated vs. 1.03 ± 0.08% control). Furthermore, P. papyraceum extract can modulate T cell subpopulations with a CD4+ effector/memory phenotype. Extract-treated cells showed increased production of IL-2 (55 ± 12 pg/mL) and IL-6 (493 ± 64 pg/mL) compared to the untreated, 21 ± 7 pg/mL and 250 ± 39 pg/mL, respectively. No significant changes were observed in the secretion of TNF-α. In addition, P. papyraceum extract displayed antibacterial activity against both Gram-negative (inhibition zone from 18.25 ± 0.50 mm to 20.28 ± 1.50 mm) and Gram-positive (inhibition zone from 10.86 ± 0.85 mm to 17.00 ± 0.82 mm) bacteria. The chemical profile of the cyanobacterial extract was determined using LC–ESI–MS/MS analysis, where at least 112 putative compounds were detected. Many of these compounds have proven different biological activities. We speculated that compounds such as betulin and the macrolide azithromycin (or their analogues) could be responsible for the immunomodulatory potential of the investigated extract. More studies are needed to determine and validate the biological activities of the determined putative compounds.
Collapse
|
14
|
Wang L, Zhang X, Wang M, Li Y, Xu J, Wei J, Li H, Ren G, Yin X. AMPD1 Is Associated With the Immune Response and Serves as a Prognostic Marker in HER2-Positive Breast Cancer. Front Oncol 2021; 11:749135. [PMID: 34900696 PMCID: PMC8660114 DOI: 10.3389/fonc.2021.749135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/25/2021] [Indexed: 01/12/2023] Open
Abstract
Background Although immunotherapy has been used in the treatment of metastatic triple negative breast cancer (TNBC), its therapeutic influence on human epidermal growth factor receptor 2 (HER2)-positive subtype remains controversial. It is therefore imperative to find biomarkers that can predict the immune response in HER2+ BC. Methods ESTIMATE was utilized to compute the ImmuneScore and StromalScore from data obtained from TCGA database, and differentially expressed genes (DEGs) were identified. In addition, univariate Cox regression was used to assess candidate genes such as AMPD1, CD33, and CCR5. Gene set enrichment analysis (GSEA) was used to further understand AMPD1-associated pathways. Moreover, immunohistochemical analyses were performed to further reveal the relationship among AMPD1, CD4 and CD8 genes. Results The expression of AMPD1 was markedly associated with disease outcome and tumor-infiltrating immune cells (TICs). In addition, AMPD1 was associated with lymph node status, age and the expression of PD-L1 and PD-L2. High AMPD1 expression was linked to longer overall survival (OS). Upregulated expression of AMPD1 correlated with the enrichment of immune-related signaling pathways. In addition, immunohistochemical analyses demonstrated a co-expression profile among AMPD1, CD4 and CD8 genes. Conclusions Taken together, our data demonstrated that AMPD1 might serve as a novel biomarker for predicting the immune response and disease outcome in HER2+ BC.
Collapse
Affiliation(s)
- Long Wang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xue Zhang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengxue Wang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunhai Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiali Xu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiaying Wei
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongzhong Li
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuedong Yin
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Tippalagama R, Singhania A, Dubelko P, Lindestam Arlehamn CS, Crinklaw A, Pomaznoy M, Seumois G, deSilva AD, Premawansa S, Vidanagama D, Gunasena B, Goonawardhana NDS, Ariyaratne D, Scriba TJ, Gilman RH, Saito M, Taplitz R, Vijayanand P, Sette A, Peters B, Burel JG. HLA-DR Marks Recently Divided Antigen-Specific Effector CD4 T Cells in Active Tuberculosis Patients. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:523-533. [PMID: 34193602 PMCID: PMC8516689 DOI: 10.4049/jimmunol.2100011] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/09/2021] [Indexed: 01/07/2023]
Abstract
Upon Ag encounter, T cells can rapidly divide and form an effector population, which plays an important role in fighting acute infections. In humans, little is known about the molecular markers that distinguish such effector cells from other T cell populations. To address this, we investigated the molecular profile of T cells present in individuals with active tuberculosis (ATB), where we expect Ag encounter and expansion of effector cells to occur at higher frequency in contrast to Mycobacterium tuberculosis-sensitized healthy IGRA+ individuals. We found that the frequency of HLA-DR+ cells was increased in circulating CD4 T cells of ATB patients, and was dominantly expressed in M. tuberculosis Ag-specific CD4 T cells. We tested and confirmed that HLA-DR is a marker of recently divided CD4 T cells upon M. tuberculosis Ag exposure using an in vitro model examining the response of resting memory T cells from healthy IGRA+ to Ags. Thus, HLA-DR marks a CD4 T cell population that can be directly detected ex vivo in human peripheral blood, whose frequency is increased during ATB disease and contains recently divided Ag-specific effector T cells. These findings will facilitate the monitoring and study of disease-specific effector T cell responses in the context of ATB and other infections.
Collapse
Affiliation(s)
- Rashmi Tippalagama
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
| | - Akul Singhania
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
| | - Paige Dubelko
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
| | | | - Austin Crinklaw
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
| | - Mikhail Pomaznoy
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
| | - Gregory Seumois
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
| | - Aruna D deSilva
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
- Faculty of Medicine, General Sir John Kotelawala Defense University, Ratmalana, Sri Lanka
| | | | | | - Bandu Gunasena
- National Hospital for Respiratory Diseases, Welisara, Sri Lanka
| | | | - Dinuka Ariyaratne
- Faculty of Medicine, General Sir John Kotelawala Defense University, Ratmalana, Sri Lanka
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Robert H Gilman
- Johns Hopkins School of Public Health, Baltimore, MD
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Mayuko Saito
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Randy Taplitz
- Department of Medicine, City of Hope National Medical Center, Duarte, CA; and
| | - Pandurangan Vijayanand
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Alessandro Sette
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Bjoern Peters
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA;
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Julie G Burel
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA;
| |
Collapse
|
17
|
Dhar C. Testing for latent tuberculosis before starting patients on immune checkpoint inhibitors. Indian J Cancer 2021; 58:469-470. [PMID: 34380834 DOI: 10.4103/ijc.ijc_283_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Chirag Dhar
- Department of Medicine and Cellular and Molecular Medicine, University of California, San Diego, U.S.A
| |
Collapse
|
18
|
Ahmed A, Rakshit S, Adiga V, Dias M, Dwarkanath P, D'Souza G, Vyakarnam A. A century of BCG: Impact on tuberculosis control and beyond. Immunol Rev 2021; 301:98-121. [PMID: 33955564 DOI: 10.1111/imr.12968] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/13/2021] [Accepted: 03/13/2021] [Indexed: 12/14/2022]
Abstract
BCG turns 100 this year and while it might not be the perfect vaccine, it has certainly contributed significantly towards eradication and prevention of spread of tuberculosis (TB). The search for newer and better vaccines for TB is an ongoing endeavor and latest results from trials of candidate TB vaccines such as M72AS01 look promising. However, recent encouraging data from BCG revaccination trials in adults combined with studies on mucosal and intravenous routes of BCG vaccination in non-human primate models have renewed interest in BCG for TB prevention. In addition, several well-demonstrated non-specific effects of BCG, for example, prevention of viral and respiratory infections, give BCG an added advantage. Also, BCG vaccination is currently being widely tested in human clinical trials to determine whether it protects against SARS-CoV-2 infection and/or death with detailed analyses and outcomes from several ongoing trials across the world awaited. Through this review, we attempt to bring together information on various aspects of the BCG-induced immune response, its efficacy in TB control, comparison with other candidate TB vaccines and strategies to improve its efficiency including revaccination and alternate routes of administration. Finally, we discuss the future relevance of BCG use especially in light of its several heterologous benefits.
Collapse
Affiliation(s)
- Asma Ahmed
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Srabanti Rakshit
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Mary Dias
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India
| | | | - George D'Souza
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India.,Department of Pulmonary Medicine, St John's Medical College, Bangalore, India
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, Guy's Hospital, King's College London, London, UK
| |
Collapse
|
19
|
Weiner J, Svetlicky N, Kang J, Sadat M, Khan K, Duttargi A, Stovroff M, Moturi S, Kara Balla A, Hyang Kwon D, Kallakury B, Hawksworth J, Subramanian S, Yazigi N, Kaufman S, Pasieka HB, Matsumoto CS, Robson SC, Pavletic S, Zasloff M, Fishbein TM, Kroemer A. CD69+ resident memory T cells are associated with graft-versus-host disease in intestinal transplantation. Am J Transplant 2021; 21:1878-1892. [PMID: 33226726 PMCID: PMC10364625 DOI: 10.1111/ajt.16405] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 09/30/2020] [Accepted: 11/13/2020] [Indexed: 01/25/2023]
Abstract
Graft-versus-host disease (GvHD) is a common, morbid complication after intestinal transplantation (ITx) with poorly understood pathophysiology. Resident memory T cells (TRM ) are a recently described CD69+ memory T cell subset localizing to peripheral tissue. We observed that T effector memory cells (TEM ) in the blood increase during GvHD and hypothesized that they derive from donor graft CD69+TRM migrating into host blood and tissue. To probe this hypothesis, graft and blood lymphocytes from 10 ITx patients with overt GvHD and 34 without were longitudinally analyzed using flow cytometry. As hypothesized, CD4+ and CD8+CD69+TRM were significantly increased in blood and grafts of GvHD patients, alongside higher cytokine and activation marker expression. The majority of CD69+TRM were donor derived as determined by multiplex immunostaining. Notably, CD8/PD-1 was significantly elevated in blood prior to transplantation in patients who later had GvHD, and percentages of HLA-DR, CD57, PD-1, and naïve T cells differed significantly between GvHD patients who died vs. those who survived. Overall, we demonstrate that (1) there were significant increases in TEM at the time of GvHD, possibly of donor derivation; (2) donor TRM in the graft are a possible source; and (3) potential biomarkers for the development and prognosis of GvHD exist.
Collapse
Affiliation(s)
- Joshua Weiner
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Nina Svetlicky
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Mohammed Sadat
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Anju Duttargi
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Merrill Stovroff
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Sangeetha Moturi
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Abdalla Kara Balla
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Dong Hyang Kwon
- Department of Pathology, MedStar Georgetown University Hospital, Washington, District of Columbia
| | - Bhaskar Kallakury
- Department of Pathology, MedStar Georgetown University Hospital, Washington, District of Columbia
| | - Jason Hawksworth
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia.,Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Sukanya Subramanian
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Nada Yazigi
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Stuart Kaufman
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Helena B Pasieka
- Division of Dermatology, MedStar Georgetown University Hospital, Georgetown University Medical Center, Washington, District of Columbia
| | - Cal S Matsumoto
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Simon C Robson
- Departments of Anesthesiology and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Steven Pavletic
- National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Michael Zasloff
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Thomas M Fishbein
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
20
|
Morgan J, Muskat K, Tippalagama R, Sette A, Burel J, Lindestam Arlehamn CS. Classical CD4 T cells as the cornerstone of antimycobacterial immunity. Immunol Rev 2021; 301:10-29. [PMID: 33751597 PMCID: PMC8252593 DOI: 10.1111/imr.12963] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/11/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Tuberculosis is a significant health problem without an effective vaccine to combat it. A thorough understanding of the immune response and correlates of protection is needed to develop a more efficient vaccine. The immune response against Mycobacterium tuberculosis (Mtb) is complex and involves all aspects of the immune system, however, the optimal protective, non‐pathogenic T cell response against Mtb is still elusive. This review will focus on discussing CD4 T cell immunity against mycobacteria and its importance in Mtb infection with a primary focus on human studies. We will in particular discuss the large heterogeneity of immune cell subsets that have been revealed by recent immunological investigations at an unprecedented level of detail. These studies have identified specific classical CD4 T cell subsets important for immune responses against Mtb in various states of infection. We further discuss the functional attributes that have been linked to the various subsets such as upregulation of activation markers and cytokine production. Another important topic to be considered is the antigenic targets of Mtb‐specific immune responses, and how antigen reactivity is influenced by both disease state and environmental exposure(s). These are key points for both vaccines and immune diagnostics development. Ultimately, these factors are holistically considered in the definition and investigations of what are the correlates on protection and resolution of disease.
Collapse
Affiliation(s)
- Jeffrey Morgan
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kaylin Muskat
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Rashmi Tippalagama
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Julie Burel
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | |
Collapse
|
21
|
Weinberg A, Tugizov S, Pandiyan P, Jin G, Rakshit S, Vyakarnam A, Naglik JR. Innate immune mechanisms to oral pathogens in oral mucosa of HIV-infected individuals. Oral Dis 2020; 26 Suppl 1:69-79. [PMID: 32862519 PMCID: PMC7570967 DOI: 10.1111/odi.13470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A crucial aspect of mucosal HIV transmission is the interaction between HIV, the local environmental milieu and immune cells. The oral mucosa comprises many host cell types including epithelial cells, CD4 + T cells, dendritic cells and monocytes/macrophages, as well as a diverse microbiome predominantly comprising bacterial species. While the oral epithelium is one of the first sites exposed to HIV through oral-genital contact and nursing infants, it is largely thought to be resistant to HIV transmission via mechanisms that are still unclear. HIV-1 infection is also associated with predisposition to secondary infections, such as tuberculosis, and other diseases including cancer. This review addresses the following questions that were discussed at the 8th World Workshop on Oral Health and Disease in AIDS held in Bali, Indonesia, 13 September –15 September 2019: (a) How does HIV infection affect epithelial cell signalling? (b) How does HIV infection affect the production of cytokines and other innate antimicrobial factors, (c) How is the mucosal distribution and function of immune cells altered in HIV infection? (d) How do T cells affect HIV (oral) pathogenesis and cancer? (e) How does HIV infection lead to susceptibility to TB infections?
Collapse
Affiliation(s)
- Aaron Weinberg
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sharof Tugizov
- Department of Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ge Jin
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Srabanti Rakshit
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Annapurna Vyakarnam
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
22
|
Vickers MA, Darboe F, Muefong CN, Mbayo G, Barry A, Gindeh A, Njie S, Riley AJ, Sarr B, Sambou B, Dockrell HM, Charalambous S, Rachow A, Owolabi O, Jayasooriya S, Sutherland JS. Monitoring Anti-tuberculosis Treatment Response Using Analysis of Whole Blood Mycobacterium tuberculosis Specific T Cell Activation and Functional Markers. Front Immunol 2020; 11:572620. [PMID: 33679684 PMCID: PMC7931252 DOI: 10.3389/fimmu.2020.572620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Background Blood-based biomarkers have been proposed as an alternative to current sputum-based treatment monitoring methods in active tuberculosis (ATB). The aim of this study was to validate previously described phenotypic, activation, and cytokine markers of treatment response in a West African cohort. Methods Whole blood immune responses to Mycobacterium tuberculosis ESAT-6/CFP-10 (EC) and purified protein derivative (PPD) were measured in twenty adults at baseline and after 2 months of standard TB treatment. Patients were classified as fast or slow responders based on a negative or positive sputum culture result at 2 months, respectively. Cellular expression of activation markers (CD38, HLA-DR), memory markers (CD27), and functional intracellular cytokine and proliferation (IFN-γ, Ki-67, TNF-α) markers were measured using multi-color flow cytometry. Results There was a significant increase in the proportion of CD4+CD27+ cells expressing CD38 and HLA-DR following EC stimulation at 2 months compared to baseline (p = 0.0328 and p = 0.0400, respectively). Following PPD stimulation, slow treatment responders had a significantly higher proportion of CD8+CD27–IFN-γ+ (p = 0.0105) and CD4+CD27+HLA-DR+CD38+ (p = 0.0077) T cells than fast responders at baseline. Receiver operating curve analysis of these subsets resulted in 80% sensitivity and 70 and 100% specificity, respectively (AUC of 0.82, p = 0.0156 and 0.84, p = 0.0102). Conclusion Our pilot data show reductions in expression of T cell activation markers were seen with treatment, but this was not associated with fast or slow sputum conversion at 2 months. However, baseline proportions of activated T cell subsets are potentially predictive of the subsequent speed of response to treatment.
Collapse
Affiliation(s)
- Molly A Vickers
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Fatoumatta Darboe
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Caleb N Muefong
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Georgetta Mbayo
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Amadou Barry
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Awa Gindeh
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Sainabou Njie
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Abi-Janet Riley
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Binta Sarr
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Basil Sambou
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Hazel M Dockrell
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Andrea Rachow
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Olumuyiwa Owolabi
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Shamanthi Jayasooriya
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia.,Academic Unit of Primary Care, University of Sheffield, Sheffield, United Kingdom
| | - Jayne S Sutherland
- Vaccines and Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| |
Collapse
|
23
|
Ahmed A, Vyakarnam A. Emerging patterns of regulatory T cell function in tuberculosis. Clin Exp Immunol 2020; 202:273-287. [PMID: 32639588 PMCID: PMC7670141 DOI: 10.1111/cei.13488] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/12/2020] [Accepted: 06/22/2020] [Indexed: 12/22/2022] Open
Abstract
Tuberculosis (TB) is one of the top 10 causes of mortality worldwide from a single infectious agent and has significant implications for global health. A major hurdle in the development of effective TB vaccines and therapies is the absence of defined immune‐correlates of protection. In this context, the role of regulatory T cells (Treg), which are essential for maintaining immune homeostasis, is even less understood. This review aims to address this knowledge gap by providing an overview of the emerging patterns of Treg function in TB. Increasing evidence from studies, both in animal models of infection and TB patients, points to the fact the role of Tregs in TB is dependent on disease stage. While Tregs might expand and delay the appearance of protective responses in the early stages of infection, their role in the chronic phase perhaps is to counter‐regulate excessive inflammation. New data highlight that this important homeostatic role of Tregs in the chronic phase of TB may be compromised by the expansion of activated human leucocyte antigen D‐related (HLA‐DR)+CD4+ suppression‐resistant effector T cells. This review provides a comprehensive and critical analysis of the key features of Treg cells in TB; highlights the importance of a balanced immune response as being important in TB and discusses the importance of probing not just Treg frequency but also qualitative aspects of Treg function as part of a comprehensive search for novel TB treatments.
Collapse
Affiliation(s)
- A Ahmed
- Laboratory of Immunology of HIV-TB Co-infection, Center for Infectious Disease Research (CIDR), Indian Institute of Science (IISc), Bangalore, India
| | - A Vyakarnam
- Laboratory of Immunology of HIV-TB Co-infection, Center for Infectious Disease Research (CIDR), Indian Institute of Science (IISc), Bangalore, India.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, Guy's Hospital, King's College London (KCL), London, UK
| |
Collapse
|
24
|
Basu S, Elkington P, Rao NA. Pathogenesis of ocular tuberculosis: New observations and future directions. Tuberculosis (Edinb) 2020; 124:101961. [PMID: 33010848 DOI: 10.1016/j.tube.2020.101961] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/22/2020] [Accepted: 06/03/2020] [Indexed: 01/01/2023]
Abstract
Ocular tuberculosis (OTB) encompasses all forms of intra- and extra-ocular inflammation associated with Mycobacterium tuberculosis (Mtb) infection. However, the organism is rarely found in ocular fluid samples of diseased eyes, rendering the pathomechanisms of the disease unclear. This confounds clinical decision-making in diagnosis and treatment of OTB. Here, we critically review existing human and animal data related to ocular inflammation and TB pathogenesis to unravel likely pathomechanisms of OTB. Broadly there appear to be two fundamental mechanisms that may underlie the development of TB-associated ocular inflammation: a. inflammatory response to live/replicating Mtb in the eye, and b. immune mediated ocular inflammation induced by non-viable Mtb or its components in the eye. This distinction is significant as in direct Mtb-driven mechanisms, diagnosis and treatment would be aimed at detection of Mtb-infection and its elimination; while indirect mechanisms would primarily require anti-inflammatory therapy with adjunctive anti-TB therapy. Further, we discuss how that most clinical phenotypes of OTB likely represent a combination of both mechanisms, with one being predominant than the other.
Collapse
Affiliation(s)
- Soumyava Basu
- Retina and Uveitis Service, L V Prasad Eye Institute (Mithu Tulsi Chanrai Campus), Bhubaneswar, India.
| | - Paul Elkington
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Narsing A Rao
- USC-Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
25
|
Ellwanger JH, Kaminski VDL, Rodrigues AG, Kulmann-Leal B, Chies JAB. CCR5 and CCR5Δ32 in bacterial and parasitic infections: Thinking chemokine receptors outside the HIV box. Int J Immunogenet 2020; 47:261-285. [PMID: 32212259 DOI: 10.1111/iji.12485] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
The CCR5 molecule was reported in 1996 as the main HIV-1 co-receptor. In that same year, the CCR5Δ32 genetic variant was described as a strong protective factor against HIV-1 infection. These findings led to extensive research regarding the CCR5, culminating in critical scientific advances, such as the development of CCR5 inhibitors for the treatment of HIV infection. Recently, the research landscape surrounding CCR5 has begun to change. Different research groups have realized that, since CCR5 has such important effects in the chemokine system, it could also affect other different physiological systems. Therefore, the effect of reduced CCR5 expression due to the presence of the CCR5Δ32 variant began to be further studied. Several studies have investigated the role of CCR5 and the impacts of CCR5Δ32 on autoimmune and inflammatory diseases, various types of cancer, and viral diseases. However, the role of CCR5 in diseases caused by bacteria and parasites is still poorly understood. Therefore, the aim of this article is to review the role of CCR5 and the effects of CCR5Δ32 on bacterial (brucellosis, osteomyelitis, pneumonia, tuberculosis and infection by Chlamydia trachomatis) and parasitic infections (toxoplasmosis, leishmaniasis, Chagas disease and schistosomiasis). Basic information about each of these infections was also addressed. The neglected role of CCR5 in fungal disease and emerging studies regarding the action of CCR5 on regulatory T cells are briefly covered in this review. Considering the "renaissance of CCR5 research," this article is useful for updating researchers who develop studies involving CCR5 and CCR5Δ32 in different infectious diseases.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Valéria de Lima Kaminski
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Andressa Gonçalves Rodrigues
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Bruna Kulmann-Leal
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| |
Collapse
|
26
|
Schoettler N, Hrusch CL, Blaine KM, Sperling AI, Ober C. Transcriptional programming and T cell receptor repertoires distinguish human lung and lymph node memory T cells. Commun Biol 2019; 2:411. [PMID: 31754641 PMCID: PMC6853923 DOI: 10.1038/s42003-019-0657-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Antigen-specific memory T cells persist for years after exposure to a pathogen and provide effective recall responses. Many memory T cell subsets have been identified and differ in abundance throughout tissues. This study focused on CD4 and CD8 memory T cells from paired human lung and lung draining lymph node (LDLN) samples and identified substantial differences in the transcriptional landscape of these subsets, including higher expression of an array of innate immune receptors in lung T cells which were further validated by flow cytometry. Using T cell receptor analysis, we determined the clonal overlap between memory T cell subsets within the lung and within the LDLN, and this was greater than the clonal overlap observed between memory T cell subsets compared across tissues. Our results suggest that lung and LDLN memory T cells originate from different precursor pools, recognize distinct antigens and likely have separate roles in immune responses.
Collapse
Affiliation(s)
- Nathan Schoettler
- Department of Medicine, The University of Chicago, Chicago, USA
- Department of Human Genetics, The University of Chicago, Chicago, USA
| | - Cara L Hrusch
- Department of Medicine, The University of Chicago, Chicago, USA
| | - Kelly M Blaine
- Department of Medicine, The University of Chicago, Chicago, USA
| | - Anne I Sperling
- Department of Medicine, The University of Chicago, Chicago, USA
| | - Carole Ober
- Department of Human Genetics, The University of Chicago, Chicago, USA
| |
Collapse
|
27
|
Shah NM, Imami N, Kelleher P, Barclay WS, Johnson MR. Pregnancy-related immune suppression leads to altered influenza vaccine recall responses. Clin Immunol 2019; 208:108254. [PMID: 31470087 DOI: 10.1016/j.clim.2019.108254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/17/2019] [Accepted: 08/26/2019] [Indexed: 01/12/2023]
Abstract
Pregnancy is a risk factor for severe influenza infection. Despite achieving seroprotective antibody titres post immunisation fewer pregnant women experience a reduction in influenza-like illness compared to non-pregnant cohorts. This may be due to the effects that immune-modulation in pregnancy has on vaccine efficacy leading to a less favourable immunologic response. To understand this, we investigated the antigen-specific cellular responses and leukocyte phenotype in pregnant and non-pregnant women who achieved seroprotection post immunisation. We show that pregnancy is associated with better antigen-specific inflammatory (IFN-γ) responses and an expansion of central memory T cells (Tcm) post immunisation, but low-level pregnancy-related immune regulation (HLA-G, PIBF) and associated reduced B-cell antibody maintenance (TGF-β) suggest poor immunologic responses compared to the non-pregnant. Thus far, studies of influenza vaccine immunogenicity have focused on the induction of antibodies but understanding additional vaccine-related cellular responses is needed to fully appreciate how pregnancy impacts on vaccine effectiveness.
Collapse
Affiliation(s)
- Nishel M Shah
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom.
| | - Nesrina Imami
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Peter Kelleher
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Wendy S Barclay
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Mark R Johnson
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| |
Collapse
|
28
|
Guo P, Bi K, Lu Z, Wang K, Xu Y, Wu H, Cao Y, Jiang H. CCR5/CCR5 ligand-induced myeloid-derived suppressor cells are related to the progression of endometriosis. Reprod Biomed Online 2019; 39:704-711. [PMID: 31427176 DOI: 10.1016/j.rbmo.2019.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/25/2019] [Accepted: 05/22/2019] [Indexed: 11/19/2022]
Abstract
RESEARCH QUESTION Immunological disorders have been reported to promote the progression of endometriosis. Several recent studies have shown that myeloid-derived suppressor cells (MDSC) drive the progression of endometriosis. The aim of this case-control study was to test whether CCR5 and its ligands drive MDSC accumulation and play a role in the progression of endometriosis. DESIGN Thirty-six endometriosis patients and 20 controls were recruited. All subjects underwent laparoscopy. An ELISA kit was used to define CCR5 ligands in plasma and peritoneal fluid from endometriosis patients; flow cytometry was then used to characterize CCR5+MDSC in peripheral blood and peritoneal fluid. RESULTS Data showed that endometriosis patients displayed a significantly higher production of plasma CCL3 (P = 0.046) and peritoneal fluid CCL3/5 (P = 0.042/0.036) compared with those from the uterine leiomyoma group. Furthermore, the concentrations of peritoneal fluid CCL5 were elevated in late stage patients compared with those from the uterine leiomyoma group. Accumulation of blood CCR5+Mo-MDSC was detected in endometriosis patients compared with those from both the ovarian dermoid cysts and uterine leiomyoma groups. Endometriosis patients also showed an elevation of CCR5+MDSC and CCR5+Mo-MDSC in peritoneal fluid samples compared with uterine leiomyoma samples. It was also found that enrichment of CCR5+MDSC (r = 0.6807; P < 0.0001) and CCR5+Mo-MDSC (r = 0.6893; P < 0.0001) were correlated with enhanced production of CCL5 in peritoneal fluid from endometriosis patients. CONCLUSIONS This study showed that CCR5 and its ligands could drive the progression of endometriosis by enhancing the accumulation of MDSC. These findings might produce a promising treatment that targets CCR5+MDSC for endometriosis patients.
Collapse
Affiliation(s)
- Peipei Guo
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, PR China
| | - Kaihuan Bi
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Zhimin Lu
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China
| | - Kangxia Wang
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, PR China
| | - Yuping Xu
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China
| | - Huan Wu
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China
| | - Yunxia Cao
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China.
| | - Huanhuan Jiang
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China.
| |
Collapse
|
29
|
Wang X, Cao X, Zhang W, Zhang L, Lu L, Li X, El‐Ashram S, Wu J, Chen C. Association of human leukocyte antigens-DQB2/DPA1/DPB1 polymorphism and pulmonary tuberculosis in the Chinese Uygur population. Mol Genet Genomic Med 2019; 7:e544. [PMID: 30600606 PMCID: PMC6418356 DOI: 10.1002/mgg3.544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/01/2018] [Accepted: 12/02/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Tuberculosis (TB) is the second-leading cause of death globally. Genetic polymorphisms in human leukocyte antigens (HLA)-DQB2, HLA-DPA1, and HLA-DPB1 may partly explain individual differences in TB susceptibility. METHODS We performed a hospital-based case-control study to assess the genetic influence of single-nucleotide polymorphisms (SNPs) in the HLA (HLA-DPA, HLA-DPB, and HLA-DQB) on the development of TB. There were 248 TB-infected cases and 340 healthy controls in this study. RESULTS The HLA-DQB2 rs7453920 genotype GG was applied as the reference group, the GA genotype was related to a considerably magnified risk of TB (GA vs. GG: adjusted OR = 1.547, 95% CI = 1.039-2.304, p = 0.032). Nevertheless, the other two SNPs were not associated with TB risk. Stratified analyses suggested that tobacco was associated with an increased risk of TB in HLA-DQB2 rs7453920 G>A. CONCLUSION These results suggested that the functional HLA-DQB2 rs7453920 G>A polymorphism may contribute to the genetic susceptibility to TB. Nevertheless, the results were based on a limited sample size, and larger well-designed studies are expected to confirm these preliminary findings.
Collapse
Affiliation(s)
- Xue Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases Cooperated by Education Ministry with Xinjiang ProvinceShihezi UniversityShiheziChina
| | - Xudong Cao
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases Cooperated by Education Ministry with Xinjiang ProvinceShihezi UniversityShiheziChina
| | - Wanjiang Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases Cooperated by Education Ministry with Xinjiang ProvinceShihezi UniversityShiheziChina
| | - Le Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases Cooperated by Education Ministry with Xinjiang ProvinceShihezi UniversityShiheziChina
| | - Lijun Lu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases Cooperated by Education Ministry with Xinjiang ProvinceShihezi UniversityShiheziChina
| | - Xinyue Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases Cooperated by Education Ministry with Xinjiang ProvinceShihezi UniversityShiheziChina
| | - Saeed El‐Ashram
- College of Life Science and EngineeringFoshan UniversityFoshanChina
- Faculty of ScienceKafrelsheikh Universitykafr El-SheikhEgypt
| | - Jiangdong Wu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases Cooperated by Education Ministry with Xinjiang ProvinceShihezi UniversityShiheziChina
| | - Chuangfu Chen
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases Cooperated by Education Ministry with Xinjiang ProvinceShihezi UniversityShiheziChina
| |
Collapse
|