1
|
Edwards CL, Engel JA, de Labastida Rivera F, Ng SS, Corvino D, Montes de Oca M, Frame TC, Chauhan SB, Singh SS, Kumar A, Wang Y, Na J, Mukhopadhyay P, Lee JS, Nylen S, Sundar S, Kumar R, Engwerda CR. A molecular signature for IL-10-producing Th1 cells in protozoan parasitic diseases. JCI Insight 2023; 8:e169362. [PMID: 37917177 PMCID: PMC10807716 DOI: 10.1172/jci.insight.169362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023] Open
Abstract
Control of visceral leishmaniasis (VL) depends on proinflammatory Th1 cells that activate infected tissue macrophages to kill resident intracellular parasites. However, proinflammatory cytokines produced by Th1 cells can damage tissues and require tight regulation. Th1 cell IL-10 production is an important cell-autologous mechanism to prevent such damage. However, IL-10-producing Th1 (type 1 regulatory; Tr1) cells can also delay control of parasites and the generation of immunity following drug treatment or vaccination. To identify molecules to target in order to alter the balance between Th1 and Tr1 cells for improved antiparasitic immunity, we compared the molecular and phenotypic profiles of Th1 and Tr1 cells in experimental VL caused by Leishmania donovani infection of C57BL/6J mice. We also identified a shared Tr1 cell protozoan signature by comparing the transcriptional profiles of Tr1 cells from mice with experimental VL and malaria. We identified LAG3 as an important coinhibitory receptor in patients with VL and experimental VL, and we reveal tissue-specific heterogeneity of coinhibitory receptor expression by Tr1 cells. We also discovered a role for the transcription factor Pbx1 in suppressing CD4+ T cell cytokine production. This work provides insights into the development and function of CD4+ T cells during protozoan parasitic infections and identifies key immunoregulatory molecules.
Collapse
Affiliation(s)
- Chelsea L. Edwards
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | | | | | - Susanna S. Ng
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Griffith University, School of Natural Sciences, Nathan, Australia
- Institute of Experimental Oncology, University of Bonn, Bonn, Germany
| | - Dillon Corvino
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute of Experimental Oncology, University of Bonn, Bonn, Germany
| | | | - Teija C.M. Frame
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | | | | | - Awnish Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Yulin Wang
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Griffith University, School of Natural Sciences, Nathan, Australia
| | - Jinrui Na
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | | | - Jason S. Lee
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Susanne Nylen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | | | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
2
|
Cavallone IN, Belda W, de Carvalho CHC, Laurenti MD, Passero LFD. New Immunological Markers in Chromoblastomycosis-The Importance of PD-1 and PD-L1 Molecules in Human Infection. J Fungi (Basel) 2023; 9:1172. [PMID: 38132773 PMCID: PMC10744586 DOI: 10.3390/jof9121172] [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: 10/24/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
The pathogenesis of chromoblastomycosis (CBM) is associated with Th2 and/or T regulatory immune responses, while resistance is associated with a Th1 response. However, even in the presence of IFN-γ, fungi persist in the lesions, and the reason for this persistence is unknown. To clarify the factors associated with pathogenesis, this study aimed to determine the polarization of the cellular immune response and the densities of cells that express markers of exhaustion in the skin of CBM patients. In the skin of patients with CBM, a moderate inflammatory infiltrate was observed, characterized primarily by the occurrence of histiocytes. Analysis of fungal density allowed us to divide patients into groups that exhibited low and high fungal densities; however, the intensity of the inflammatory response was not related to mycotic loads. Furthermore, patients with CBM exhibited a significant increase in the number of CD4+ and CD8+ cells associated with a high density of IL-10-, IL-17-, and IFN-γ-producing cells, indicating the presence of a chronic and mixed cellular immune response, which was also independent of fungal load. A significant increase in the number of PD-1+ and PD-L1+ cells was observed, which may be associated with the maintenance of the fungus in the skin and the progression of the disease.
Collapse
Affiliation(s)
- Italo N. Cavallone
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil;
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School, São Paulo University, São Paulo 01246-903, Brazil
| | - Walter Belda
- Dermatology Department, Medical School, São Paulo University, Clinics Hospital, São Paulo 05403-000, Brazil; (W.B.J.); (C.H.C.d.C.); (M.D.L.)
| | - Caroline Heleno C. de Carvalho
- Dermatology Department, Medical School, São Paulo University, Clinics Hospital, São Paulo 05403-000, Brazil; (W.B.J.); (C.H.C.d.C.); (M.D.L.)
| | - Marcia D. Laurenti
- Dermatology Department, Medical School, São Paulo University, Clinics Hospital, São Paulo 05403-000, Brazil; (W.B.J.); (C.H.C.d.C.); (M.D.L.)
| | - Luiz Felipe D. Passero
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil;
- Institute for Advanced Studies of Ocean (IEAMAR), São Paulo State University (UNESP), Rua João Francisco Bensdorp, 1178, São Vicente 11350-011, Brazil
| |
Collapse
|
3
|
Nagai K, Goto Y. Parasitomimetics: Can We Utilize Parasite-Derived Immunomodulatory Molecules for Interventions to Immunological Disorders? Front Immunol 2022; 13:824695. [PMID: 35386686 PMCID: PMC8977410 DOI: 10.3389/fimmu.2022.824695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/28/2022] [Indexed: 11/17/2022] Open
Abstract
Because our immune system has ability to expel microorganisms invading our body, parasites need evolution to maintain their symbiosis with the hosts. One such strategy of the parasites is to manipulate host immunity by producing immunomodulatory molecules and the ability of parasites to regulate host immunity has long been a target of research. Parasites can not only manipulate host immune response specific to them, but also influence the host's entire immune system. Such ability of the parasites may sometimes bring benefit to the hosts as many studies have indicated the "hygiene hypothesis" that a decreased opportunity of parasitic infections is associated with an increased incidence of allergy and autoimmune diseases. In other words, elucidating the mechanisms of parasites to regulate host immunity could be applied not only to resolution of parasitic infections but also to treatment of non-parasitic immunological disorders. In this review, we show how much progress has been made in the research on immunomodulation of host immunity by parasites. Here, we define the word 'parasitomimetics' as emulation of parasites' immunomodulatory systems to solve immunological problems in humans and discuss potential applications of parasite-derived molecules to other diseases.
Collapse
Affiliation(s)
| | - Yasuyuki Goto
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Seth A, Kar S. Host-directed antileishmanial interventions: Harvesting unripe fruits to reach fruition. Int Rev Immunol 2022; 42:217-236. [PMID: 35275772 DOI: 10.1080/08830185.2022.2047670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Leishmaniasis is an exemplary paradigm of immune evasion, fraught with the perils of limited clinical assistance, escalating costs of treatment and made worse with the lack of suitable vaccine. While drugs remain central to large-scale disease control, the growing emergence of parasite resistance necessitates the need for combination therapy involving host-directed immunological agents. Also, since prolonged disease progression is associated with strong immune suppression of the host, augmentation of host immunity via restoration of the immunoregulatory circuit involving antigen-presenting cells and T-cells, activation of macrophage function and/or CD4+ T helper 1 cell differentiation may serve as an ideal approach to resolve severe cases of leishmaniasis. As such, therapies that embody a synergistic approach that involve direct killing of the parasite in addition to elevating host immunity are likely to pave the way for widespread elimination of leishmaniasis in the future. With this review, we aim to recapitulate the various immunotherapeutic agents found to hold promise in antileishmanial treatment both in vitro and in vivo. These include parasite-specific antigens, dendritic cell-targeted therapy, recombinant inhibitors of various components intrinsic to immune cell signaling and agonists or antagonists to immune cells and cytokines. We also summarize their abilities to direct therapeutic skewing of the host cell-immune response and review their potential to combat the disease either alone, or as adjunct modalities.
Collapse
Affiliation(s)
- Anuradha Seth
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Susanta Kar
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
5
|
Dey NS, Senaratne S, Somaratne V, Madarasinghe NP, Seneviratne B, Forrester S, Montes de Oca M, Reis LC, Moulik S, Walrad PB, Chatterjee M, Goto H, Wickremasinghe R, Lagos D, Kaye PM, Ranasinghe S. Early reduction in PD-L1 expression predicts faster treatment response in human cutaneous leishmaniasis. J Clin Invest 2021; 131:142765. [PMID: 34609968 PMCID: PMC8592550 DOI: 10.1172/jci142765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is caused by Leishmania donovani in Sri Lanka. Pentavalent antimonials (e.g., sodium stibogluconate [SSG]) remain first-line drugs for CL with no new effective treatments emerging. We studied whole blood and lesion transcriptomes from Sri Lankan patients with CL at presentation and during SSG treatment. From lesions but not whole blood, we identified differential expression of immune-related genes, including immune checkpoint molecules, after onset of treatment. Using spatial profiling and RNA-FISH, we confirmed reduced expression of programmed death-ligand 1 (PD-L1) and indoleamine 2,3-dioxygenase 1 (IDO1) proteins on treatment in lesions of a second validation cohort and further demonstrated significantly higher expression of these checkpoint molecules on parasite-infected compared with noninfected lesional CD68+ monocytes and macrophages. Crucially, early reduction in PD-L1 but not IDO1 expression was predictive of rate of clinical cure (HR = 4.88) and occurred in parallel with reduction in parasite load. Our data support a model whereby the initial anti-leishmanial activity of antimonial drugs alleviates checkpoint inhibition on T cells, facilitating immune-drug synergism and clinical cure. Our findings demonstrate that PD-L1 expression can be used as a predictor of rapidity of clinical response to SSG treatment in Sri Lanka and support further evaluation of PD-L1 as a host-directed therapeutic in leishmaniasis.
Collapse
Affiliation(s)
- Nidhi S Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Sujai Senaratne
- Department of Parasitology, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Vijani Somaratne
- Dermatology Unit, District General Hospital Embilipitiya, Embilipitiya, Sri Lanka
| | | | - Bimalka Seneviratne
- Deparment of Pathology, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Sarah Forrester
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Marcela Montes de Oca
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Luiza Campos Reis
- Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Srija Moulik
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Pegine B Walrad
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Hiro Goto
- Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil.,Departamento de Medicina Preventiva, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Renu Wickremasinghe
- Department of Parasitology, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Dimitris Lagos
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Shalindra Ranasinghe
- Department of Parasitology, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| |
Collapse
|
6
|
Garcia de Moura R, Covre LP, Fantecelle CH, Gajardo VAT, Cunha CB, Stringari LL, Belew AT, Daniel CB, Zeidler SVV, Tadokoro CE, de Matos Guedes HL, Zanotti RL, Mosser D, Falqueto A, Akbar AN, Gomes DCO. PD-1 Blockade Modulates Functional Activities of Exhausted-Like T Cell in Patients With Cutaneous Leishmaniasis. Front Immunol 2021; 12:632667. [PMID: 33767700 PMCID: PMC7985249 DOI: 10.3389/fimmu.2021.632667] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/20/2021] [Indexed: 01/17/2023] Open
Abstract
Patients infected by Leishmania braziliensis develop debilitating skin lesions. The role of inhibitory checkpoint receptors (ICRs) that induce T cell exhaustion during this disease is not known. Transcriptional profiling identified increased expression of ICRs including PD-1, PDL-1, PDL-2, TIM-3, and CTLA-4 in skin lesions of patients that was confirmed by immunohistology where there was increased expression of PD-1, TIM-3, and CTLA-4 in both CD4+ and CD8+ T cell subsets. Moreover, PDL-1/PDL-2 ligands were increased on skin macrophages compared to healthy controls. The proportions PD1+, but not TIM-3 or CTLA-4 expressing T cells in the circulation were positively correlated with those in the lesions of the same patients, suggesting that PD-1 may regulate T cell function equally in both compartments. Blocking PD-1 signaling in circulating T cells enhanced their proliferative capacity and IFN-γ production, but not TNF-α secretion in response to L. braziliensis recall antigen challenge in vitro. While we previously showed a significant correlation between the accumulation of senescent CD8+CD45RA+CD27- T cells in the circulation and skin lesion size in the patients, there was no such correlation between the extent of PD-1 expression by circulating on T cells and the magnitude of skin lesions suggesting that exhausted-like T cells may not contribute to the cutaneous immunopathology. Nevertheless, we identified exhausted-like T cells in both skin lesions and in the blood. Targeting this population by PD-1 blockade may improve T cell function and thus accelerate parasite clearance that would reduce the cutaneous pathology in cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Renan Garcia de Moura
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitoria, Brazil
| | - Luciana Polaco Covre
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitoria, Brazil,Division of Medicine, University College London, London, United Kingdom
| | | | | | - Carla Baroni Cunha
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitoria, Brazil
| | | | - Ashton Trey Belew
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| | | | | | | | - Herbert Leonel de Matos Guedes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - David Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| | - Aloisio Falqueto
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitoria, Brazil
| | - Arne N. Akbar
- Division of Medicine, University College London, London, United Kingdom
| | - Daniel Claudio Oliveira Gomes
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitoria, Brazil,Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitoria, Brazil,*Correspondence: Daniel Claudio Oliveira Gomes,
| |
Collapse
|
7
|
Künzli M, Reuther P, Pinschewer DD, King CG. Opposing effects of T cell receptor signal strength on CD4 T cells responding to acute versus chronic viral infection. eLife 2021; 10:61869. [PMID: 33684030 PMCID: PMC7943189 DOI: 10.7554/elife.61869] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
A hallmark of adaptive immunity is CD4 T cells’ ability to differentiate into specialized effectors. A long-standing question is whether T cell receptor (TCR) signal strength can dominantly instruct the development of Th1 and T follicular helper (Tfh) cells across distinct infectious contexts. We characterized the differentiation of murine CD4 TCR transgenic T cells responding to altered peptide ligand lymphocytic choriomeningitis viruses (LCMV) derived from acute and chronic parental strains. We found that TCR signal strength exerts opposite and hierarchical effects on the balance of Th1 and Tfh cells responding to acute versus persistent infection. TCR signal strength correlates positively with Th1 generation during acute but negatively during chronic infection. Weakly activated T cells express lower levels of markers associated with chronic T cell stimulation and may resist functional inactivation. We anticipate that the panel of recombinant viruses described herein will be valuable for investigating a wide range of CD4 T cell responses.
Collapse
Affiliation(s)
- Marco Künzli
- Immune Cell Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Peter Reuther
- Division of Experimental Virology, Department of Biomedicine - Haus Petersplatz, University of Basel, Basel, Switzerland
| | - Daniel D Pinschewer
- Division of Experimental Virology, Department of Biomedicine - Haus Petersplatz, University of Basel, Basel, Switzerland
| | - Carolyn G King
- Immune Cell Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
8
|
Pérez-Antón E, Egui A, Thomas MC, Carrilero B, Simón M, López-Ruz MÁ, Segovia M, López MC. A proportion of CD4+ T cells from patients with chronic Chagas disease undergo a dysfunctional process, which is partially reversed by benznidazole treatment. PLoS Negl Trop Dis 2021; 15:e0009059. [PMID: 33539379 PMCID: PMC7888659 DOI: 10.1371/journal.pntd.0009059] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 02/17/2021] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Background Signs of senescence and the late stages of differentiation associated with the more severe forms of Chagas disease have been described in the Trypanosoma cruzi antigen-specific CD4+ T-cell population. However, the mechanisms involved in these functions are not fully known. To date, little is known about the possible impact of benznidazole treatment on the T. cruzi-specific functional response of CD4+ T cells. Methodology/Principal findings The functional capacity of CD4+ T cells was analyzed by cytometric assays in chronic Chagas disease patients, with indeterminate form (IND) and cardiac alterations (CCC) (25 and 15, respectively) before and after benznidazole treatment. An increase in the multifunctional capacity (expression of IFN-γ, IL-2, TNF-α, perforin and/or granzyme B) of the antigen-specific CD4+ T cells was observed in indeterminate versus cardiac patients, which was associated with the reduced coexpression of inhibitory receptors (2B4, CD160, CTLA-4, PD-1 and/or TIM-3). The functional profile of these cells shows statistically significant differences between IND and CCC (p<0.001), with a higher proportion of CD4+ T cells coexpressing 2 and 3 molecules in IND (54.4% versus 23.1% and 4.1% versus 2.4%, respectively). A significant decrease in the frequencies of CD4+ T cells that coexpress 2, 3 and 4 inhibitory receptors was observed in IND after 24–48 months of treatment (p<0.05, p<0.01 and p<0.05, respectively), which was associated with an increase in antigen-specific multifunctional activity. The IND group showed, at 9–12 months after treatment, an increase in the CD4+ T cell subset coproducing three molecules, which were mainly granzyme B+, perforin+ and IFN-γ+ (1.4% versus 4.5%). Conclusions/Significance A CD4+ T cell dysfunctional process was detected in chronic Chagas disease patients, being more exacerbated in those patients with cardiac symptoms. After short-term benznidazole treatment (9–12 months), indeterminate patients showed a significant increase in the frequency of multifunctional antigen-specific CD4+ T cells. Trypanosoma cruzi infection triggers several immune mechanisms in the host that do not result in a total clearance of the parasite, the persistence of which leads to the chronicity of Chagas disease. The mechanisms by which some chronic patients remain asymptomatic or become symptomatic are not entirely clear. The aim of the present manuscript is to study the CD4+ T cell population and its functional capacity in patients with different forms of chronic disease. The obtained results indicate that cells from indeterminate patients have an enhanced multifunctional profile, which is associated with the reduced expression of inhibitory molecules. CD4+ T cells from chronic patients with cardiac alterations show lower functional activity against specific antigens of the parasite and increased coexpression of inhibitory molecules. After benznidazole treatment, antigen-specific CD4+ T cells, especially those from indeterminate patients, are more likely to show a multifunctional profile and a decline in the coexpression of inhibitory receptors. These results allow us to make progress in clarifying the mechanisms that may influence disease progression and to realize the importance of antiparasitic treatment for the enhancement of the activity of the immune system.
Collapse
Affiliation(s)
- Elena Pérez-Antón
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas; Granada, Spain
| | - Adriana Egui
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas; Granada, Spain
| | - M. Carmen Thomas
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas; Granada, Spain
| | - Bartolomé Carrilero
- Unidad Regional de Medicina Tropical, Hospital Virgen de la Arrixaca; El Palmar, Murcia, Spain
| | - Marina Simón
- Unidad Regional de Medicina Tropical, Hospital Virgen de la Arrixaca; El Palmar, Murcia, Spain
| | | | - Manuel Segovia
- Unidad Regional de Medicina Tropical, Hospital Virgen de la Arrixaca; El Palmar, Murcia, Spain
| | - Manuel Carlos López
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas; Granada, Spain
- * E-mail:
| |
Collapse
|
9
|
Bogdan C. Macrophages as host, effector and immunoregulatory cells in leishmaniasis: Impact of tissue micro-environment and metabolism. Cytokine X 2020; 2:100041. [PMID: 33604563 PMCID: PMC7885870 DOI: 10.1016/j.cytox.2020.100041] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Leishmania are protozoan parasites that predominantly reside in myeloid cells within their mammalian hosts. Monocytes and macrophages play a central role in the pathogenesis of all forms of leishmaniasis, including cutaneous and visceral leishmaniasis. The present review will highlight the diverse roles of macrophages in leishmaniasis as initial replicative niche, antimicrobial effectors, immunoregulators and as safe hideaway for parasites persisting after clinical cure. These multiplex activities are either ascribed to defined subpopulations of macrophages (e.g., Ly6ChighCCR2+ inflammatory monocytes/monocyte-derived dendritic cells) or result from different activation statuses of tissue macrophages (e.g., macrophages carrying markers of of classical [M1] or alternative activation [M2]). The latter are shaped by immune- and stromal cell-derived cytokines (e.g., IFN-γ, IL-4, IL-10, TGF-β), micro milieu factors (e.g., hypoxia, tonicity, amino acid availability), host cell-derived enzymes, secretory products and metabolites (e.g., heme oxygenase-1, arginase 1, indoleamine 2,3-dioxygenase, NOS2/NO, NOX2/ROS, lipids) as well as by parasite products (e.g., leishmanolysin/gp63, lipophosphoglycan). Exciting avenues of current research address the transcriptional, epigenetic and translational reprogramming of macrophages in a Leishmania species- and tissue context-dependent manner.
Collapse
Key Words
- (L)CL, (localized) cutaneous leishmaniasis
- AHR, aryl hydrocarbon receptor
- AMP, antimicrobial peptide
- Arg, arginase
- Arginase
- CAMP, cathelicidin-type antimicrobial peptide
- CR, complement receptor
- DC, dendritic cells
- DCL, diffuse cutaneous leishmaniasis
- HO-1, heme oxygenase 1
- Hypoxia
- IDO, indoleamine-2,3-dioxygenase
- IFN, interferon
- IFNAR, type I IFN (IFN-α/β) receptor
- IL, interleukin
- Interferon-α/β
- Interferon-γ
- JAK, Janus kinase
- LPG, lipophosphoglycan
- LRV1, Leishmania RNA virus 1
- Leishmaniasis
- Macrophages
- Metabolism
- NCX1, Na+/Ca2+ exchanger 1
- NFAT5, nuclear factor of activated T cells 5
- NK cell, natural killer cell
- NO, nitric oxide
- NOS2 (iNOS), type 2 (or inducible) nitric oxide synthase
- NOX2, NADPH oxidase 2 (gp91 or cytochrome b558 β-subunit of Phox)
- Nitric oxide
- OXPHOS, mitochondrial oxidative phosphorylation
- PKDL, post kala-azar dermal leishmaniasis
- Phagocyte NADPH oxidase
- Phox, phagocyte NADPH oxidase
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SOCS, suppressor of cytokine signaling
- STAT, signal transducer and activator of transcription
- TGF-β, transforming growth factor-beta
- TLR, toll-like receptor
- Th1 (Th2), type 1 (type2) T helper cell
- Tonicity
- VL, visceral leishmaniasis
- mTOR, mammalian/mechanistic target of rapamycin
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut - klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
10
|
Pessenda G, da Silva JS. Arginase and its mechanisms in Leishmania persistence. Parasite Immunol 2020; 42:e12722. [PMID: 32294247 DOI: 10.1111/pim.12722] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/14/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Leishmaniasis is a neglected infectious disease with clinical presentations ranging from asymptomatic or mild symptoms to chronic infection and eventual death. The mechanisms of disease susceptibility and pathology have been extensively studied, but there are no steadfast rules regarding leishmaniasis. A Th1 response is usually associated with infection control, while a predominant Th2 response is detrimental to the patient. In this scenario, the enzymes arginase and inducible nitric oxide synthase represent two possible pathways of immune response. While the former contributes to parasite replication, the latter is crucial for its control. In the present review, we collected study results that associate arginase expression in patients and in experimental models with disease susceptibility/chronicity and show some proposed mechanisms that explain the role of arginase in maintaining Leishmania infection, including polyamine and thiol synthesis, tissue-resident macrophage (TRM) proliferation and activation and T-cell suppression and exhaustion.
Collapse
Affiliation(s)
- Gabriela Pessenda
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Santana da Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fundação Oswaldo Cruz Bi-institucional, Ribeirão Preto, Brazil
| |
Collapse
|
11
|
Leth S, Jensen-Fangel S. Programmed cell death protein 1 (PD-1) in infection. APMIS 2020; 128:177-187. [PMID: 32304591 DOI: 10.1111/apm.13045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022]
Abstract
Exhausted and dysfunctional T cells triggered by infection and cancer render the immune system unable to eliminate these pathogens. Pharmacologic blockade of the surface receptors that inhibit T-cell function has shown remarkable success in patients with various malignancies. In this Review, we discuss the emerging evidence of inhibiting checkpoint pathways as a potential role in controlling or clearing infectious diseases. Though interesting tendencies, much work is still needed in order to develop safe strategies that can be translated into clinically relevant outcomes in patients with infections.
Collapse
Affiliation(s)
- Steffen Leth
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Jensen-Fangel
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
12
|
Jubel JM, Barbati ZR, Burger C, Wirtz DC, Schildberg FA. The Role of PD-1 in Acute and Chronic Infection. Front Immunol 2020; 11:487. [PMID: 32265932 PMCID: PMC7105608 DOI: 10.3389/fimmu.2020.00487] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/03/2020] [Indexed: 12/26/2022] Open
Abstract
PD-1 as an immune checkpoint molecule down-regulates T cell activity during immune responses in order to prevent autoimmune tissue damage. In chronic infections or tumors, lasting antigen-exposure leads to permanent PD-1 expression that can limit immune-mediated clearance of pathogens or degenerated cells. Blocking PD-1 can enhance T cell function; in cancer treatment PD-1 blockade is already used as a successful therapy. However, the role of PD-1 expression and blocking in the context of acute and chronic infections is less defined. Building on its success in cancer therapy leads to the hypothesis that blocking PD-1 in infectious diseases is also beneficial in acute or chronic infections. This review will focus on the role of PD-1 expression in acute and chronic infections with virus, bacteria, and parasites, with a particular focus on recent studies regarding PD-1 blockade in infectious diseases.
Collapse
Affiliation(s)
- Jil M Jubel
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
13
|
da Fonseca-Martins AM, Ramos TD, Pratti JES, Firmino-Cruz L, Gomes DCO, Soong L, Saraiva EM, de Matos Guedes HL. Immunotherapy using anti-PD-1 and anti-PD-L1 in Leishmania amazonensis-infected BALB/c mice reduce parasite load. Sci Rep 2019; 9:20275. [PMID: 31889072 PMCID: PMC6937231 DOI: 10.1038/s41598-019-56336-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/04/2019] [Indexed: 01/07/2023] Open
Abstract
Leishmaniasis is a neglected disease, for which current treatment presents numerous issues. Leishmania amazonensis is the etiological agent of cutaneous and diffuse cutaneous leishmaniasis. The roles of the programmed death-1 (PD-1) receptor on lymphocytes and its ligand (PD-L1) on antigen-presenting cells have been well studied in tumor and other infection models; but little is known about their roles in non-healing cutaneous leishmaniasis. In this study, we observed that L. amazonensis induced PD-1 expression on both CD4+ and CD8+ T cells and PD-L1 on dendritic cells on BALB/c mice. We tested the therapeutic potential of anti-PD-1 and anti-PD-L1 monoclonal antibodies (MoAbs) against a non-healing L. amazonensis infection in BALB/c mice, and that anti-PD-1 and anti-PD-L1 treatment significantly increased IFN-γ-producing CD4+ and CD8+ T cells, respectively. Compared with infection controls, mice treated with anti-PD-1 and anti-PD-L1, but not anti-PD-L2, displayed bigger lesions with significantly lower parasite loads. Treatment did not affect anti-Leishmania antibody (IgM, IgG, IgG1 and IgG2a) or IL-10 production, but anti-PD-1 treatment reduced both IL-4 and TGF-β production. Together, our results highlight the therapeutic potential of an anti-PD-1-based treatment in promoting the reinvigoration of T cells for the control of parasite burden.
Collapse
Affiliation(s)
- Alessandra M da Fonseca-Martins
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Paulo de Góes Microbiology Institute, Immunology Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tadeu D Ramos
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana E S Pratti
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luan Firmino-Cruz
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Lynn Soong
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Elvira M Saraiva
- Paulo de Góes Microbiology Institute, Immunology Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Herbert L de Matos Guedes
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Imunofarmacologia, Grupo de Imunologia e Vacinologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,Universidade Federal do Rio de Janeiro - Campus Duque de Caxias Professor Geraldo Cidade, Rio de Janeiro, RJ, Brazil. .,Laboratório Interdisciplinar de pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Abers MS, Lionakis MS, Kontoyiannis DP. Checkpoint Inhibition and Infectious Diseases: A Good Thing? Trends Mol Med 2019; 25:1080-1093. [PMID: 31494023 DOI: 10.1016/j.molmed.2019.08.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022]
Abstract
The mammalian immune system has evolved the capacity to detect and destroy tumor cells. Tumors utilize multiple strategies to evade host immune surveillance, including the induction of the checkpoint molecules cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) to suppress antitumor immunity. Pharmacologic blockade of these molecules with checkpoint inhibitors (CPIs) restores T cell function and prolongs survival in patients with various malignancies. Emerging evidence suggests that the same checkpoint pathways may play a crucial role during infections. Indeed, CPIs appear promising as immunotherapeutic agents in infectious diseases, although their efficacy varies depending on pathogen-, cell-, and organ-specific factors. More research will be necessary to clarify the effects and safety of CPIs on clinically relevant outcomes of human infection.
Collapse
Affiliation(s)
- Michael S Abers
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center (UT-MDACC), Houston, TX, USA
| |
Collapse
|
15
|
Paduch K, Debus A, Rai B, Schleicher U, Bogdan C. Resolution of Cutaneous Leishmaniasis and Persistence of Leishmania major in the Absence of Arginase 1. THE JOURNAL OF IMMUNOLOGY 2019; 202:1453-1464. [PMID: 30665936 DOI: 10.4049/jimmunol.1801249] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/16/2018] [Indexed: 01/29/2023]
Abstract
Arginase (Arg) 1 is expressed by hematopoietic (e.g., macrophages) and nonhematopoietic cells (e.g., endothelial cells) and converts l-arginine into ornithine and urea. The enzyme is implicated in tissue repair but also antagonizes the production of NO by type 2 NO synthase in myeloid cells and thereby impedes the control of intracellular parasites such as Leishmania major In this study, we tested whether Arg1 is required for spontaneous healing of acute cutaneous leishmaniasis in C57BL/6 mice and for lifelong parasite persistence in draining lymph nodes (dLNs) of healed mice. In vitro, bone marrow-derived macrophages and lymphoid endothelial cells readily expressed Arg1 in response to IL-4 and/or IL-13, whereas skin or dLN fibroblasts failed to do so, even during hypoxia. In vivo, Arg1 was found in skin lesions and, to a much lower extent, also in dLNs of acutely infected C57BL/6 mice but became undetectable at both sites after healing. Deletion of Arg1 in hematopoietic and endothelial cells using Tie2Cre+/-Arg1fl/fl C57BL/6 mice abolished the expression of Arg1 in skin lesions and dLNs but did not affect development and resolution of skin lesions, parasite burden, NO production, or host cell tropism of L. major during the acute or persistent phase of infection. Similar to wild-type controls, parasites persisting in Arg1-deficient mice favored NO synthase 2-negative areas and mainly resided in myeloid cells and fibroblasts. We conclude that Arg1 expression by hematopoietic and endothelial cells is completely dispensable for clinical resolution of cutaneous leishmaniasis and for long-term persistence of L. major.
Collapse
Affiliation(s)
- Katrin Paduch
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and
| | - Andrea Debus
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and
| | - Baplu Rai
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
16
|
Egui A, Ledesma D, Pérez-Antón E, Montoya A, Gómez I, Robledo SM, Infante JJ, Vélez ID, López MC, Thomas MC. Phenotypic and Functional Profiles of Antigen-Specific CD4 + and CD8 + T Cells Associated With Infection Control in Patients With Cutaneous Leishmaniasis. Front Cell Infect Microbiol 2018; 8:393. [PMID: 30510917 PMCID: PMC6252334 DOI: 10.3389/fcimb.2018.00393] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/19/2018] [Indexed: 01/03/2023] Open
Abstract
The host immunological response is a key factor determining the pathogenesis of cutaneous leishmaniasis. It is known that a Th1 cellular response is associated with infection control and that antigen-specific memory T cells are necessary for the development of a rapid and strong protective cellular response. The present manuscript reports the analysis of the functional and phenotypic profiles of antigen-specific CD4+ and CD8+ T cells from patients cured of cutaneous leishmaniasis (CL), patients with an active process of cutaneous leishmaniasis, asymptomatic individuals with a positive Montenegro test and healthy donors (HD). Peripheral blood mononuclear cells (PBMCs) from the patients exhibited a lymphoproliferative capacity after stimulation with total soluble protein from either Leishmania panamensis (SLpA) or Leishmania infantum (SLiA) or with a recombinant paraflagellar rod protein-1 (rPFR1). Higher frequencies of antigen-specific TNAIVE cells, mainly following stimulation with rPFR1, were observed in asymptomatic and cured patients than in patients with active cutaneous leishmaniasis, while T cells from patients with active cutaneous leishmaniasis showed a higher percentage of effector memory T cells (TEM for CD4+ T cells and TEMRA for CD8+ T cells). The amount of antigen-specific CD57+/CD8+ TEMRA cells in patients with active cutaneous leishmaniasis was higher than that in cured patients and asymptomatic subjects. Regarding functionality, a more robust multifunctional CD8+ T cell response was detected in cured patients than in those with active cutaneous leishmaniasis. Moreover, cured patients showed a significant increase in the frequency of cells expressing a Th1-type cytotoxic production profile (IFN-γ+/granzyme-B/+perforin+). Patients with an active leishmaniosis process had a significantly higher frequency of CD8+ T cells expressing the inhibitory CD160 and 2B4 receptors than did cured patients. The expression profile observed in cured patients could be indicative of an imbalance toward a CD8+ Th1 response, which could be associated with infection control; consequently, the determination of this profile could be a useful tool for facilitating the clinical follow-up of patients with cutaneous leishmaniasis. The results also suggest a possible exhaustion process of CD8+ T cells associated with the evolution of Leishmania infection.
Collapse
Affiliation(s)
- Adriana Egui
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Darién Ledesma
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Elena Pérez-Antón
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Andrés Montoya
- Programa de Estudio y Control de Enfermedades Tropicales, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Inmaculada Gómez
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Sara María Robledo
- Programa de Estudio y Control de Enfermedades Tropicales, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Juan José Infante
- Bionaturis Group, Bioorganic Research and Services, S.A., Jerez de la Frontera, Spain
| | - Ivan Darío Vélez
- Programa de Estudio y Control de Enfermedades Tropicales, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Manuel C López
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - M Carmen Thomas
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
17
|
Singh RK, Gannavaram S, Ismail N, Kaul A, Gedda MR, Nakhasi HL. Centrin-Deleted Leishmania donovani Parasites Help CD4 + T Cells to Acquire Th1 Phenotype and Multi-Functionality Through Downregulation of CD200-CD200R Immune Inhibitory Axis. Front Immunol 2018; 9:1176. [PMID: 29915577 PMCID: PMC5994488 DOI: 10.3389/fimmu.2018.01176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/11/2018] [Indexed: 11/26/2022] Open
Abstract
The protozoan parasite Leishmania has evolved several strategies to undermine host defense mechanisms by inducing Th2-type adaptive immunity and suppressing effector functions of Th1 phenotype. In our earlier studies, using centrin gene-deleted Leishmania (LdCen−/−) parasites as an immunogen, we have shown induction of an effective Th1-type immunity and robust memory responses that mediate protection against virulent challenge. However, role of inhibitory signals in Leishmania vaccine induced immunity in general, and LdCen−/− in particular has not been studied. Herein, we report that immunization with LdCen−/− parasites produces more functional Th1-type CD4+ T cells via downregulation of CD200–CD200R immune inhibitory axis compared to wild-type infection. We found that expression of CD200 and CD200R was significantly reduced in LdCen−/− infection compared to wild-type infection. Diminished CD200–CD200R signaling in LdCen−/− infection enabled proliferation of CD4+ T cells and resulted in the induction of pro-inflammatory cytokines and suppression of anti-inflammatory response. The effects of diminished CD200–CD200R signaling by LdCen−/− were most evident in the suppression of IL-10-producing CD4+ T cells that helped enhance more Th1 cytokine producing and multi-functional T cells compared to wild-type infection. In vivo blocking of CD200 expression with anti-CD200 treatment in wild-type infected mice limited Th2 response as indicated by reduction of IL-10-producing Tr1 cells and reduced parasite burden. On the other hand, treatment with anti-CD200 improved the LdCen−/− vaccine-induced multifunctional response and reduction in splenic parasite load upon challenge. Taken together, these studies demonstrate the role of CD200–CD200R signals in the protection induced by LdCen−/− parasites.
Collapse
Affiliation(s)
- Rakesh K Singh
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States.,Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Nevien Ismail
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Amit Kaul
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States.,Johns Hopkins Medical Institution, Johns Hopkins University, Baltimore, MD, United States
| | - Mallikarjuna Rao Gedda
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
18
|
STAT3 regulates cytotoxicity of human CD57+ CD4+ T cells in blood and lymphoid follicles. Sci Rep 2018; 8:3529. [PMID: 29476109 PMCID: PMC5824848 DOI: 10.1038/s41598-018-21389-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/10/2018] [Indexed: 11/09/2022] Open
Abstract
A subset of human follicular helper T cells (TFH) cells expresses CD57 for which no distinct function has been identified. We show that CD57+ TFH cells are universally PD-1hi, but compared to their CD57- PD-1hi counterparts, express little IL-21 or IL-10 among others. Instead, CD57 expression on TFH cells marks cytotoxicity transcriptional signatures that translate into only a weak cytotoxic phenotype. Similarly, circulating PD-1+ CD57+ CD4+ T cells make less cytokine than their CD57- PD-1+ counterparts, but have a prominent cytotoxic phenotype. By analysis of responses to STAT3-dependent cytokines and cells from patients with gain- or loss-of-function STAT3 mutations, we show that CD4+ T cell cytotoxicity is STAT3-dependent. TFH formation also requires STAT3, but paradoxically, once formed, PD-1hi cells become unresponsive to STAT3. These findings suggest that changes in blood and germinal center cytotoxicity might be affected by changes in STAT3 signaling, or modulation of PD-1 by therapy.
Collapse
|
19
|
Abstract
The upregulation of immune checkpoint molecules, such as programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte antigen 4 (CTLA4), on immune cells occurs during acute infections, such as malaria, as well as during chronic persistent viral infections, including HIV and hepatitis B virus. These pathways are important for preventing immune-driven pathology but can also limit immune-mediated clearance of the infection. The recent success of immune checkpoint blockade in cancer therapy suggests that targeting these pathways would also be effective for preventing and treating a range of infectious diseases. Here, we review our current understanding of immune checkpoint pathways in the pathogenesis of infectious diseases and discuss the potential for therapeutically targeting these pathways in this setting.
Collapse
Affiliation(s)
- Michelle N Wykes
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3000, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
20
|
Filippis C, Arens K, Noubissi Nzeteu GA, Reichmann G, Waibler Z, Crauwels P, van Zandbergen G. Nivolumab Enhances In Vitro Effector Functions of PD-1 + T-Lymphocytes and Leishmania-Infected Human Myeloid Cells in a Host Cell-Dependent Manner. Front Immunol 2017; 8:1880. [PMID: 29312350 PMCID: PMC5743744 DOI: 10.3389/fimmu.2017.01880] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/11/2017] [Indexed: 12/19/2022] Open
Abstract
Functional impairment of T-cells and a concomitant augmented expression of programmed death-1 (PD-1) have been observed in visceral leishmaniasis patients, as well as in experimental models for visceral and cutaneous leishmaniasis. The PD-1/PD-1-ligand (PD-1/PD-L) interaction negatively regulates T-cell effector functions, which are required for parasite control during leishmaniasis. The aim of this study was to elucidate the impact of the PD-1/PD-L axis in a human primary in vitro infection model of Leishmania major (Lm). Blocking the PD-1/PD-L interaction with nivolumab increased T-cell proliferation and release of the proinflammatory cytokines TNFα and IFNγ during the cocultivation of Lm-infected human monocyte-derived macrophages (hMDMs) or dendritic cells (hMDDC) with autologous PD-1+-lymphocytes. As a consequence Lm infection decreased, being the most pronounced in hMDDC, compared to proinflammatory hMDM1 and anti-inflammatory hMDM2. Focusing on hMDDC, we could partially reverse effects mediated by PD-1 blockade by neutralizing TNFα but not by neutralizing IFNγ. Furthermore, PD-1 blockade increased intracellular expression of perforin, granulysin, and granzymes in proliferating CD4+-T-cells, which might be implicated in reduction of Lm-infected cells. In all, our data describe an important role for the PD-1/PD-L axis upon Lm infection using a human primary cell system. These data contribute to a better understanding of the PD-1-induced T-cell impairment during disease and its influence on immune effector mechanisms to combat Lm infection.
Collapse
Affiliation(s)
| | - Katharina Arens
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | | | | | - Zoe Waibler
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Peter Crauwels
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Ger van Zandbergen
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany.,Immunology, Johannes Gutenberg University of Mainz, Mainz, Germany
| |
Collapse
|
21
|
Leptin regulates Granzyme-A, PD-1 and CTLA-4 expression in T cell to control visceral leishmaniasis in BALB/c Mice. Sci Rep 2017; 7:14664. [PMID: 29116252 PMCID: PMC5676676 DOI: 10.1038/s41598-017-15288-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/24/2017] [Indexed: 01/01/2023] Open
Abstract
Visceral leishmaniasis (VL) is responsible for several deaths in malnourished children accompanied by diminished circulating leptin and impaired cell-mediated immunity. Typically, leptin deficiency is associated with the Th2 polarization that markedly coincides with the pathogenesis of VL. The aim of the present study was to unravel the prophylactic role of leptin in malnutrition-coupled VL mice. Interestingly, we observed that L. donovani infection itself reduces the serum leptin levels in malnutrition. Exogenous leptin restored severe body weight loss and parasite load in the spleen and liver of malnourished infected mice compared to controls. Leptin increases functional CD8+ T-cell population, Granzyme-A expression down-regulates anergic T-cell markers such as PD-1 and CTLA-4. It was also noticed that, leptin suppresses GM-CSF mRNA expression in parasite favored monocytes and reduced arginase activity in bone marrow derived macrophage indicate macrophages dependent T-cell activation and proliferation. Leptin-induced IFN-γ, IL-2, and TNF-α cytokines in the culture supernatant of splenocytes upon soluble leishmanial antigen (SLA) stimulation and significantly up-regulates serum IgG2a titers, which help to generate Th1 immune response in VL. Furthermore, leptin induced a granulomatous response and restored L. donovani induced tissue degeneration in the liver. Altogether, our findings suggest the exogenous leptin can restore T cell mediated immunity in malnourished VL mice.
Collapse
|
22
|
Hammami A, Abidin BM, Charpentier T, Fabié A, Duguay AP, Heinonen KM, Stäger S. HIF-1α is a key regulator in potentiating suppressor activity and limiting the microbicidal capacity of MDSC-like cells during visceral leishmaniasis. PLoS Pathog 2017; 13:e1006616. [PMID: 28892492 PMCID: PMC5608422 DOI: 10.1371/journal.ppat.1006616] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/21/2017] [Accepted: 08/29/2017] [Indexed: 12/20/2022] Open
Abstract
Leishmania donovani is known to induce myelopoiesis and to dramatically increase extramedullary myelopoiesis. This results in splenomegaly, which is then accompanied by disruption of the splenic microarchitecture, a chronic inflammatory environment, and immunosuppression. Chronically inflamed tissues are typically hypoxic. The role of hypoxia on myeloid cell functions during visceral leishmaniasis has not yet been studied. Here we show that L. donovani promotes the output from the bone marrow of monocytes with a regulatory phenotype that function as safe targets for the parasite. We also demonstrate that splenic myeloid cells acquire MDSC-like function in a HIF-1α-dependent manner. HIF-1α is also involved in driving the polarization towards M2-like macrophages and rendering intermediate stage monocytes more susceptible to L. donovani infection. Our results suggest that HIF-1α is a major player in the establishment of chronic Leishmania infection and is crucial for enhancing immunosuppressive functions and lowering leishmanicidal capacity of myeloid cells. The protozoan parasite Leishmania donovani causes chronic infection in the spleen, which is accompanied by a chronic inflammatory environment, an enlargement of the organ, and immunosuppression. The environment of chronically inflamed tissues is characterized by low oxygen levels and tissue disruption, which induce the expression of the transcription factor HIF-1α in all cells. The kinetics of monocyte production and differentiation in the bone marrow and the spleen, and the role of hypoxia in myeloid cell functions during visceral leishmaniasis have not yet been studied. Here we show that L. donovani promotes the output from the bone marrow of monocytes with a regulatory phenotype that function as safe targets for the parasite. We also demonstrate that HIF-1α potentiates inhibitory functions of myeloid cells and is involved in driving the polarization towards M2-like macrophages and rendering them more susceptible to L. donovani infection. Our results suggest that HIF-1α is a major player in the establishment of chronic Leishmania infection and is crucial for enhancing immunosuppressive functions and lowering leishmanicidal capacity of myeloid cells.
Collapse
Affiliation(s)
- Akil Hammami
- INRS-Institut Armand-Frappier and Center for Host-Parasite interactions, 531 Boulevard des Prairies, Laval (QC), Canada
| | - Belma Melda Abidin
- INRS-Institut Armand-Frappier and Center for Host-Parasite interactions, 531 Boulevard des Prairies, Laval (QC), Canada
| | - Tania Charpentier
- INRS-Institut Armand-Frappier and Center for Host-Parasite interactions, 531 Boulevard des Prairies, Laval (QC), Canada
| | - Aymeric Fabié
- INRS-Institut Armand-Frappier and Center for Host-Parasite interactions, 531 Boulevard des Prairies, Laval (QC), Canada
| | - Annie-Pier Duguay
- INRS-Institut Armand-Frappier and Center for Host-Parasite interactions, 531 Boulevard des Prairies, Laval (QC), Canada
| | - Krista M. Heinonen
- INRS-Institut Armand-Frappier and Center for Host-Parasite interactions, 531 Boulevard des Prairies, Laval (QC), Canada
| | - Simona Stäger
- INRS-Institut Armand-Frappier and Center for Host-Parasite interactions, 531 Boulevard des Prairies, Laval (QC), Canada
- * E-mail:
| |
Collapse
|
23
|
Badirzadeh A, Taheri T, Taslimi Y, Abdossamadi Z, Heidari-Kharaji M, Gholami E, Sedaghat B, Niyyati M, Rafati S. Arginase activity in pathogenic and non-pathogenic species of Leishmania parasites. PLoS Negl Trop Dis 2017; 11:e0005774. [PMID: 28708893 PMCID: PMC5529023 DOI: 10.1371/journal.pntd.0005774] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/26/2017] [Accepted: 07/05/2017] [Indexed: 12/25/2022] Open
Abstract
Proliferation of Leishmania (L.) parasites depends on polyamine availability, which can be generated by the L-arginine catabolism and the enzymatic activity of arginase (ARG) of the parasites and of the mammalian hosts. In the present study, we characterized and compared the arginase (arg) genes from pathogenic L. major and L. tropica and from non-pathogenic L. tarentolae. We quantified the level of the ARG activity in promastigotes and macrophages infected with pathogenic L. major and L. tropica and non-pathogenic L. tarentolae amastigotes. The ARG's amino acid sequences of the pathogenic and non-pathogenic Leishmania demonstrated virtually 98.6% and 88% identities with the reference L. major Friedlin ARG. Higher ARG activity was observed in all pathogenic promastigotes as compared to non-pathogenic L. tarentolae. In vitro infection of human macrophage cell line (THP1) with pathogenic and non-pathogenic Leishmania spp. resulted in increased ARG activities in the infected macrophages. The ARG activities present in vivo were assessed in susceptible BALB/c and resistant C57BL/6 mice infected with L. major, L. tropica and L. tarentolae. We demonstrated that during the development of the infection, ARG is induced in both strains of mice infected with pathogenic Leishmania. However, in L. major infected BALB/c mice, the induction of ARG and parasite load increased simultaneously according to the time course of infection, whereas in C57BL/6 mice, the enzyme is upregulated solely during the period of footpad swelling. In L. tropica infected mice, the footpads' swellings were slow to develop and demonstrated minimal cutaneous pathology and ARG activity. In contrast, ARG activity was undetectable in mice inoculated with the non-pathogenic L. tarentolae. Our data suggest that infection by Leishmania parasites can increase ARG activity of the host and provides essential polyamines for parasite salvage and its replication. Moreover, the ARG of Leishmania is vital for parasite proliferation and required for infection in mice. ARG activity can be used as one of the main marker of the disease severity.
Collapse
Affiliation(s)
- Alireza Badirzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tahereh Taheri
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Abdossamadi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Heidari-Kharaji
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Gholami
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Baharehsadat Sedaghat
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Niyyati
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
24
|
Solana JC, Ramírez L, Corvo L, de Oliveira CI, Barral-Netto M, Requena JM, Iborra S, Soto M. Vaccination with a Leishmania infantum HSP70-II null mutant confers long-term protective immunity against Leishmania major infection in two mice models. PLoS Negl Trop Dis 2017; 11:e0005644. [PMID: 28558043 PMCID: PMC5466331 DOI: 10.1371/journal.pntd.0005644] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 06/09/2017] [Accepted: 05/16/2017] [Indexed: 12/24/2022] Open
Abstract
Background The immunization with genetically attenuated Leishmania cell lines has been associated to the induction of memory and effector T cell responses against Leishmania able to control subsequent challenges. A Leishmania infantum null mutant for the HSP70-II genes has been described, possessing a non-virulent phenotype. Methodology/Principal findings The L. infantum attenuated parasites (LiΔHSP70-II) were inoculated in BALB/c (intravenously and subcutaneously) and C57BL/6 (subcutaneously) mice. An asymptomatic infection was generated and parasites diminished progressively to become undetectable in most of the analyzed organs. However, inoculation resulted in the long-term induction of parasite specific IFN-γ responses able to control the disease caused by a challenge of L. major infective promastigotes. BALB/c susceptible mice showed very low lesion development and a drastic decrease in parasite burdens in the lymph nodes draining the site of infection and internal organs. C57BL/6 mice did not show clinical manifestation of disease, correlated to the rapid migration of Leishmania specific IFN-γ producing T cells to the site of infection. Conclusion/Significance Inoculation of the LiΔHSP70-II attenuated line activates mammalian immune system for inducing moderate pro-inflammatory responses. These responses are able to confer long-term protection in mice against the infection of L. major virulent parasites. Despite numerous efforts made, a vaccine against leishmaniasis for humans is not available. Attempts based on parasite fractions or selected antigens failed to confer long lasting protection. On the other side, leishmanization, which consists in the inoculation of live virulent parasites in hidden parts of the body, is effective against cutaneous leishmaniasis in humans but objectionable in terms of biosafety. Some efforts have been made to design live vaccines to make leishmanization safer. A promising strategy is the development of genetically attenuated parasites, able to confer immunity without undesirable side effects. Here, we have employed an attenuated L. infantum line (LiΔHSP70-II) as a vaccine against heterologous challenge with L. major in two experimental models. Infection with LiΔHSP70-II parasites does not cause pathology and induces long-term protection based on the induction of IFN-γ producing T cells that are recruited rapidly and specifically to the site of challenge with the virulent parasites. These results support the idea of using attenuated parasites for vaccination.
Collapse
Affiliation(s)
- José Carlos Solana
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Ramírez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Corvo
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Manoel Barral-Netto
- Centro de Pesquisas Gonçalo Moniz (Fundação Oswaldo Cruz-FIOCRUZ). Salvador, Bahia, Brazil
| | - José María Requena
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Salvador Iborra
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail: (SI); (MS)
| | - Manuel Soto
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail: (SI); (MS)
| |
Collapse
|
25
|
Hwang S, Cobb DA, Bhadra R, Youngblood B, Khan IA. Blimp-1-mediated CD4 T cell exhaustion causes CD8 T cell dysfunction during chronic toxoplasmosis. J Exp Med 2016; 213:1799-818. [PMID: 27481131 PMCID: PMC4995081 DOI: 10.1084/jem.20151995] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/03/2016] [Indexed: 12/26/2022] Open
Abstract
CD8, but not CD4, T cells are considered critical for control of chronic toxoplasmosis. Although CD8 exhaustion has been previously reported in Toxoplasma encephalitis (TE)-susceptible model, our current work demonstrates that CD4 not only become exhausted during chronic toxoplasmosis but this dysfunction is more pronounced than CD8 T cells. Exhausted CD4 population expressed elevated levels of multiple inhibitory receptors concomitant with the reduced functionality and up-regulation of Blimp-1, a transcription factor. Our data demonstrates for the first time that Blimp-1 is a critical regulator for CD4 T cell exhaustion especially in the CD4 central memory cell subset. Using a tamoxifen-dependent conditional Blimp-1 knockout mixed bone marrow chimera as well as an adoptive transfer approach, we show that CD4 T cell-intrinsic deletion of Blimp-1 reversed CD8 T cell dysfunction and resulted in improved pathogen control. To the best of our knowledge, this is a novel finding, which demonstrates the role of Blimp-1 as a critical regulator of CD4 dysfunction and links it to the CD8 T cell dysfunctionality observed in infected mice. The critical role of CD4-intrinsic Blimp-1 expression in mediating CD4 and CD8 T cell exhaustion may provide a rational basis for designing novel therapeutic approaches.
Collapse
Affiliation(s)
- SuJin Hwang
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC 20037
| | - Dustin A Cobb
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC 20037 Department of Microbiology, Immunology, and Cancer Biology, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908
| | - Rajarshi Bhadra
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC 20037
| | - Ben Youngblood
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Imtiaz A Khan
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC 20037
| |
Collapse
|
26
|
Mandell MA, Beverley SM. Concomitant Immunity Induced by Persistent Leishmania major Does Not Preclude Secondary Re-Infection: Implications for Genetic Exchange, Diversity and Vaccination. PLoS Negl Trop Dis 2016; 10:e0004811. [PMID: 27352043 PMCID: PMC4924822 DOI: 10.1371/journal.pntd.0004811] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/07/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Many microbes have evolved the ability to co-exist for long periods of time within other species in the absence of overt pathology. Evolutionary biologists have proposed benefits to the microbe from 'asymptomatic persistent infections', most commonly invoking increased likelihood of transmission by longer-lived hosts. Typically asymptomatic persistent infections arise from strong containment by the immune system, accompanied by protective immunity; such 'vaccination' from overt disease in the presence of a non-sterilizing immune response is termed premunition or concomitant immunity. Here we consider another potential benefit of persistence and concomitant immunity to the parasite: the 'exclusion' of competing super-infecting strains, which would favor transmission of the original infecting organism. METHODOLOGY / PRINCIPLE FINDINGS To investigate this in the protozoan parasite Leishmania major, a superb model for the study of asymptomatic persistence, we used isogenic lines of comparable virulence bearing independent selectable markers. One was then used to infect genetically resistant mice, yielding infections which healed and progressed to asymptomatic persistent infection; these mice were then super-infected with the second marked line. As anticipated, super-infection yielded minimal pathology, showing that protective immunity against disease pathology had been established. The relative abundance of the primary and super-infecting secondary parasites was then assessed by plating on selective media. The data show clearly that super-infecting parasites were able to colonize the immune host effectively, achieving numbers comparable to and sometimes greater than that of the primary parasite. CONCLUSIONS / SIGNIFICANCE We conclude that induction of protective immunity does not guarantee the Leishmania parasite exclusive occupation of the infected host. This finding has important consequences to the maintenance and generation of parasite diversity in the natural Leishmania infectious cycle alternating between mammalian and sand fly hosts.
Collapse
Affiliation(s)
- Michael A. Mandell
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
27
|
Gannavaram S, Bhattacharya P, Ismail N, Kaul A, Singh R, Nakhasi HL. Modulation of Innate Immune Mechanisms to Enhance Leishmania Vaccine-Induced Immunity: Role of Coinhibitory Molecules. Front Immunol 2016; 7:187. [PMID: 27242794 PMCID: PMC4865500 DOI: 10.3389/fimmu.2016.00187] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/02/2016] [Indexed: 12/14/2022] Open
Abstract
No licensed human vaccines are currently available against any parasitic disease including leishmaniasis. Several antileishmanial vaccine formulations have been tested in various animal models, including genetically modified live-attenuated parasite vaccines. Experimental infection studies have shown that Leishmania parasites utilize a broad range of strategies to undermine effector properties of host phagocytic cells, i.e., dendritic cells (DCs) and macrophages (MΦ). Furthermore, Leishmania parasites have evolved strategies to actively inhibit TH1 polarizing functions of DCs and to condition the infected MΦ toward anti-inflammatory/alternative/M2 phenotype. The altered phenotype of phagocytic cells is characterized by decreased production of antimicrobial reactive oxygen, nitrogen molecules, and pro-inflammatory cytokines, such as IFN-γ, IL-12, and TNF-α. These early events limit the activation of TH1-effector cells and set the stage for pathogenesis. Furthermore, this early control of innate immunity by the virulent parasites results in substantial alteration in the adaptive immunity characterized by reduced proliferation of CD4+ and CD8+ T cells and TH2-biased immunity that results in production of anti-inflammatory cytokines, such as TGF-β, and IL-10. More recent studies have also documented the induction of coinhibitory ligands, such as CTLA-4, PD-L1, CD200, and Tim-3, that induce exhaustion and/or non-proliferation in antigen-experienced T cells. Most of these studies focus on viral infections in chronic phase, thus limiting the direct application of these results to parasitic infections and much less to parasitic vaccines. However, these studies suggest that vaccine-induced protective immunity can be modulated using strategies that enhance the costimulation that might reduce the threshold necessary for T cell activation and conversely by strategies that reduce or block inhibitory molecules, such as PD-L1 and CD200. In this review, we will focus on the polarization of antigen-presenting cells and subsequent role of costimulatory and coinhibitory molecules in mediating vaccine-induced immunity using live-attenuated Leishmania parasites as specific examples.
Collapse
Affiliation(s)
- Sreenivas Gannavaram
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| | - Parna Bhattacharya
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| | - Nevien Ismail
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| | - Amit Kaul
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| | - Rakesh Singh
- Department of Biochemistry, Banaras Hindu University , Varanasi , India
| | - Hira L Nakhasi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Food and Drug Administration , Silver Spring, MD , USA
| |
Collapse
|
28
|
Holowka T, Castilho TM, Garcia AB, Sun T, McMahon-Pratt D, Bucala R. Leishmania-encoded orthologs of macrophage migration inhibitory factor regulate host immunity to promote parasite persistence. FASEB J 2016; 30:2249-65. [PMID: 26956417 DOI: 10.1096/fj.201500189r] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/12/2016] [Indexed: 11/11/2022]
Abstract
Leishmania major encodes 2 orthologs of the cytokine macrophage migration inhibitory factor (MIF), whose functions in parasite growth or in the host-parasite interaction are unknown. To determine the importance of Leishmania-encoded MIF, both LmMIF genes were removed to produce an mif(-/-) strain of L. major This mutant strain replicated normally in vitro but had a 2-fold increased susceptibility to clearance by macrophages. Mice infected with mif(-/-) L. major, when compared to the wild-type strain, also showed a 3-fold reduction in parasite burden. Microarray and functional analyses revealed a reduced ability of mif(-/-) L. major to activate antigen-presenting cells, resulting in a 2-fold reduction in T-cell priming. In addition, there was a reduction in inflammation and effector CD4 T-cell formation in mif(-/-) L. major-infected mice when compared to mice infected with wild-type L. major Notably, effector CD4 T cells that developed during infection with mif(-/-) L. major demonstrated statistically significant differences in markers of functional exhaustion, including increased expression of IFN-γ and IL-7R, reduced expression of programmed death-1, and decreased apoptosis. These data support a role for LmMIF in promoting parasite persistence by manipulating the host response to increase the exhaustion and depletion of protective CD4 T cells.-Holowka, T., Castilho, T. M., Baeza Garcia, A., Sun, T., McMahon-Pratt, D., Bucala, R. Leishmania-encoded orthologs of macrophage migration inhibitory factor regulate host immunity to promote parasite persistence.
Collapse
Affiliation(s)
- Thomas Holowka
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; and
| | - Tiago M Castilho
- Department of Epidemiology of Microbial Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Alvaro Baeza Garcia
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; and
| | - Tiffany Sun
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; and
| | - Diane McMahon-Pratt
- Department of Epidemiology of Microbial Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; and Department of Epidemiology of Microbial Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
29
|
La X, Zhang F, Li Y, Li J, Guo Y, Zhao H, Pang N, Ma X, Wen H, Fan H, Ding J. Upregulation of PD-1 on CD4⁺CD25⁺ T cells is associated with immunosuppression in liver of mice infected with Echinococcus multilocularis. Int Immunopharmacol 2015; 26:357-66. [PMID: 25907244 DOI: 10.1016/j.intimp.2015.04.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/25/2015] [Accepted: 04/07/2015] [Indexed: 12/18/2022]
Abstract
Alveolar echinococcosis is a zoonotic disease caused by Echinococcus multilocularis (E. multilocularis) infection. The relationship between PD-1/PD-L1 pathway and Tregs at different stages of E. multilocularis infection has rarely been reported. This study aims to investigate the role of PD-1/PD-L1 in immunosuppression of Tregs in E. multilocularis infection. Hematoxylin-eosin staining, flow cytometry, immunohistochemistry, quantitative RT-PCR analysis, cytometric bead array and MTT assay were used to analyze liver pathological changes, percentages of PD-1(+) Tregs and PD-L1(+) dendritic cells (DCs), expression levels of PD-1, PD-L1 and Foxp3, levels of interleukin-10 (IL-10) and transforming growth factor beta (TGF-β) and proliferation of lymphocytes. During middle-late stage (day 30 to day 330) the percentages of PD-1(+) Tregs and PD-L1(+) DCs together with levels of Foxp3, IL-10 and TGF-β increased significantly and maintained at high level. The expression of PD-1 and PD-L1 was increased with the enlarging erosion of E. multilocularis, and was mainly distributed in hepatic sinus, fibrous wall of alveolar hydatid and germinal layer around foci of infection. PD-1/PD-L1 promoted the secretion of IL-10 and TGF-β. Our results indicate that engagement of the PD-1 and PD-L1 correlates with inhibition of T-cell effector function, cytokine secretion and proliferation. High expression of PD-1/PD-L1 may play an important role in stimulating CD4(+)CD25(+) T cells, and maintaining peripheral tolerance and immune evasion during chronic infection of E. multilocularis.
Collapse
Affiliation(s)
- Xiaolin La
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Fengbo Zhang
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Yanhua Li
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Jun Li
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Yuyuan Guo
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Hui Zhao
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Nannan Pang
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Xiumin Ma
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Hao Wen
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China
| | - Haining Fan
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Qinghai University, 251 Xining Road, 810000, Xi-ning, Qinghai, China.
| | - Jianbing Ding
- Xinjiang Laboratory of Hydatid Fundamental Medicine, First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, 830011,Urumqi, Xinjiang, China; Department of Immunology, School of Preclinical Medicine of Xinjiang Medical University 393 Xinyi Road, 830011, Urumqi, Xinjiang, China.
| |
Collapse
|
30
|
T cell exhaustion and Interleukin 2 downregulation. Cytokine 2015; 71:339-47. [DOI: 10.1016/j.cyto.2014.11.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 01/30/2023]
|
31
|
Braun NA, Celada LJ, Herazo-Maya JD, Abraham S, Shaginurova G, Sevin CM, Grutters J, Culver DA, Dworski R, Sheller J, Massion PP, Polosukhin VV, Johnson JE, Kaminski N, Wilkes DS, Oswald-Richter KA, Drake WP. Blockade of the programmed death-1 pathway restores sarcoidosis CD4(+) T-cell proliferative capacity. Am J Respir Crit Care Med 2014; 190:560-71. [PMID: 25073001 DOI: 10.1164/rccm.201401-0188oc] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
RATIONALE Effective therapeutic interventions for chronic, idiopathic lung diseases remain elusive. Normalized T-cell function is an important contributor to spontaneous resolution of pulmonary sarcoidosis. Up-regulation of inhibitor receptors, such as programmed death-1 (PD-1) and its ligand, PD-L1, are important inhibitors of T-cell function. OBJECTIVES To determine the effects of PD-1 pathway blockade on sarcoidosis CD4(+) T-cell proliferative capacity. METHODS Gene expression profiles of sarcoidosis and healthy control peripheral blood mononuclear cells were analyzed at baseline and follow-up. Flow cytometry was used to measure ex vivo expression of PD-1 and PD-L1 on systemic and bronchoalveolar lavage-derived cells of subjects with sarcoidosis and control subjects, as well as the effects of PD-1 pathway blockade on cellular proliferation after T-cell receptor stimulation. Immunohistochemistry analysis for PD-1/PD-L1 expression was conducted on sarcoidosis, malignant, and healthy control lung specimens. MEASUREMENTS AND MAIN RESULTS Microarray analysis demonstrates longitudinal increase in PDCD1 gene expression in sarcoidosis peripheral blood mononuclear cells. Immunohistochemistry analysis revealed increased PD-L1 expression within sarcoidosis granulomas and lung malignancy, but this was absent in healthy lungs. Increased numbers of sarcoidosis PD-1(+) CD4(+) T cells are present systemically, compared with healthy control subjects (P < 0.0001). Lymphocytes with reduced proliferative capacity exhibited increased proliferation with PD-1 pathway blockade. Longitudinal analysis of subjects with sarcoidosis revealed reduced PD-1(+) CD4(+) T cells with spontaneous clinical resolution but not with disease progression. CONCLUSIONS Analogous to the effects in other chronic lung diseases, these findings demonstrate that the PD-1 pathway is an important contributor to sarcoidosis CD4(+) T-cell proliferative capacity and clinical outcome. Blockade of the PD-1 pathway may be a viable therapeutic target to optimize clinical outcomes.
Collapse
|
32
|
Tran LS, Bergot AS, Mattarollo SR, Mittal D, Frazer IH. Human papillomavirus e7 oncoprotein transgenic skin develops an enhanced inflammatory response to 2,4-dinitrochlorobenzene by an arginase-1-dependent mechanism. J Invest Dermatol 2014; 134:2438-2446. [PMID: 24732401 PMCID: PMC4134683 DOI: 10.1038/jid.2014.186] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 03/05/2014] [Accepted: 03/21/2014] [Indexed: 11/09/2022]
Abstract
We have shown that the expression of human papillomavirus type 16 E7 (HPV16.E7) protein within epithelial cells results in local immune suppression and a weak and ineffective immune response to E7 similar to that occuring in HPV-associated premalignancy and cancers. However, a robust acute inflammatory stimulus can overcome this to enable immune elimination of HPV16.E7-transformed epithelial cells. 2,4-Dinitrochlorobenzene (DNCB) can elicit acute inflammation and it has been shown to initiate the regression of HPV-associated genital warts. Although the clinical use of DNCB is discouraged owing to its mutagenic potential, understanding how DNCB-induced acute inflammation alters local HPV16.E7-mediated immune suppression might lead to better treatments. Here, we show that topical DNCB application to skin expressing HPV16.E7 as a transgene induces a hyperinflammatory response, which is not seen in nontransgenic control animals. The E7-associated inflammatory response is characterized by enhanced expression of Th2 cytokines and increased infiltration of CD11b(+)Gr1(int)F4/80(+)Ly6C(hi)Ly6G(low) myeloid cells, producing arginase-1. Inhibition of arginase with an arginase-specific inhibitor, N(omega)-hydroxy-nor-L-arginine, ameliorates the DNCB-induced inflammatory response. Our results demonstrate that HPV16.E7 protein enhances DNCB-associated production of arginase-1 by myeloid cells and consequent inflammatory cellular infiltration of skin.
Collapse
Affiliation(s)
- L S Tran
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - A-S Bergot
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - S R Mattarollo
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - D Mittal
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - I H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| |
Collapse
|
33
|
Rodrigues V, Cordeiro-da-Silva A, Laforge M, Ouaissi A, Akharid K, Silvestre R, Estaquier J. Impairment of T cell function in parasitic infections. PLoS Negl Trop Dis 2014; 8:e2567. [PMID: 24551250 PMCID: PMC3923671 DOI: 10.1371/journal.pntd.0002567] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In mammals subverted as hosts by protozoan parasites, the latter and/or the agonists they release are detected and processed by sensors displayed by many distinct immune cell lineages, in a tissue(s)-dependent context. Focusing on the T lymphocyte lineage, we review our present understanding on its transient or durable functional impairment over the course of the developmental program of the intracellular parasites Leishmania spp., Plasmodium spp., Toxoplasma gondii, and Trypanosoma cruzi in their mammalian hosts. Strategies employed by protozoa to down-regulate T lymphocyte function may act at the initial moment of naïve T cell priming, rendering T cells anergic or unresponsive throughout infection, or later, exhausting T cells due to antigen persistence. Furthermore, by exploiting host feedback mechanisms aimed at maintaining immune homeostasis, parasites can enhance T cell apoptosis. We will discuss how infections with prominent intracellular protozoan parasites lead to a general down-regulation of T cell function through T cell anergy and exhaustion, accompanied by apoptosis, and ultimately allowing pathogen persistence.
Collapse
Affiliation(s)
- Vasco Rodrigues
- CNRS FRE 3235, Université Paris Descartes, Paris, France
- Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Anabela Cordeiro-da-Silva
- Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto, Porto, Portugal
| | | | - Ali Ouaissi
- Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Khadija Akharid
- Département de Biologie, Faculté des Sciences Aîn-Chock, Université Hassan II-Casablanca, Casablanca, Maroc
| | - Ricardo Silvestre
- Parasite Disease Group, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- * E-mail: (RS); (JE)
| | - Jérôme Estaquier
- CNRS FRE 3235, Université Paris Descartes, Paris, France
- Université Laval, Centre de Recherche en Infectiologie, Québec, Canada
- * E-mail: (RS); (JE)
| |
Collapse
|
34
|
Abstract
Parasite-driven dysfunctional adaptive immunity represents an emerging hypothesis to explain the chronic or persistent nature of parasitic infections, as well as the observation that repeated exposure to most parasitic organisms fails to engender sterilizing immunity. This review discusses recent examples from clinical studies and experimental models of parasitic infection that substantiate the role for immune dysfunction in the inefficient generation and maintenance of potent anti-parasitic immunity. Better understanding of the complex interplay between parasites, host adaptive immunity, and relevant negative regulatory circuits will inform efforts to enhance resistance to chronic parasitic infections through vaccination or immunotherapy.
Collapse
Affiliation(s)
- Ryan A Zander
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 ; Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Noah S Butler
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 ; Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| |
Collapse
|
35
|
Abstract
Parasitic diseases caused by helminth and protozoan infections remain one of the largest global public health problems for mankind. While natural immunity in man is rare or slow to develop for many parasites, the immune response is capable of recognizing and responding to infection by utilizing a number of different immunological mechanisms. This special topics journal issue examines many of the key findings in the recent literature regarding the immune response against helminth and protozoan infections, as well as highlighting areas in which our current knowledge falls short. The question of how we can tailor immune responses to prevent or reduce disease burden is a burning question within the field of immunoparasitology.
Collapse
Affiliation(s)
- Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - P'ng Loke
- Department of Microbiology, New York University School of Medicine, New York, NY 10010
| |
Collapse
|
36
|
dos Reis MBG, Manjolin LC, Maquiaveli CDC, Santos-Filho OA, da Silva ER. Inhibition of Leishmania (Leishmania) amazonensis and rat arginases by green tea EGCG, (+)-catechin and (-)-epicatechin: a comparative structural analysis of enzyme-inhibitor interactions. PLoS One 2013; 8:e78387. [PMID: 24260115 PMCID: PMC3832641 DOI: 10.1371/journal.pone.0078387] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/17/2013] [Indexed: 11/21/2022] Open
Abstract
Epigallocatechin-3-gallate (EGCG), a dietary polyphenol (flavanol) from green tea, possesses leishmanicidal and antitrypanosomal activity. Mitochondrial damage was observed in Leishmania treated with EGCG, and it contributed to the lethal effect. However, the molecular target has not been defined. In this study, EGCG, (+)-catechin and (−)-epicatechin were tested against recombinant arginase from Leishmania amazonensis (ARG-L) and rat liver arginase (ARG-1). The compounds inhibit ARG-L and ARG-1 but are more active against the parasite enzyme. Enzyme kinetics reveal that EGCG is a mixed inhibitor of the ARG-L while (+)-catechin and (−)-epicatechin are competitive inhibitors. The most potent arginase inhibitor is (+)-catechin (IC50 = 0.8 µM) followed by (−)-epicatechin (IC50 = 1.8 µM), gallic acid (IC50 = 2.2 µM) and EGCG (IC50 = 3.8 µM). Docking analyses showed different modes of interaction of the compounds with the active sites of ARG-L and ARG-1. Due to the low IC50 values obtained for ARG-L, flavanols can be used as a supplement for leishmaniasis treatment.
Collapse
Affiliation(s)
- Matheus Balduíno Goncalves dos Reis
- Programa de Iniciação Científica da Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, São Paulo, Brazil
| | - Letícia Correa Manjolin
- Programa de Iniciação Científica da Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, São Paulo, Brazil
| | - Claudia do Carmo Maquiaveli
- Programa de pós-graduação em Fisiologia, Departamento de Fisiologia, Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Osvaldo Andrade Santos-Filho
- Laboratório de Modelagem Molecular, Departamento de Síntese Orgânica, Farmanguinhos/Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edson Roberto da Silva
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
37
|
Esch KJ, Juelsgaard R, Martinez PA, Jones DE, Petersen CA. Programmed death 1-mediated T cell exhaustion during visceral leishmaniasis impairs phagocyte function. THE JOURNAL OF IMMUNOLOGY 2013; 191:5542-50. [PMID: 24154626 DOI: 10.4049/jimmunol.1301810] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Control of Leishmania infantum infection is dependent upon Th1 CD4(+) T cells to promote macrophage intracellular clearance of parasites. Deficient CD4(+) T cell effector responses during clinical visceral leishmaniasis (VL) are associated with elevated production of IL-10. In the primary domestic reservoir of VL, dogs, we define occurrence of both CD4(+) and CD8(+) T cell exhaustion as a significant stepwise loss of Ag-specific proliferation and IFN-γ production, corresponding to increasing VL symptoms. Exhaustion was associated with a 4-fold increase in the population of T cells with surface expression of programmed death 1 (PD-1) between control and symptomatic populations. Importantly, exhausted populations of CD8(+) T cells and to a lesser extent CD4(+) T cells were present prior to onset of clinical VL. VL-exhausted T cells did not undergo significant apoptosis ex vivo after Ag stimulation. Ab block of PD-1 ligand, B7.H1, promoted return of CD4(+) and CD8(+) T cell function and dramatically increased reactive oxygen species production in cocultured monocyte-derived phagocytes. As a result, these phagocytes had decreased parasite load. To our knowledge, we demonstrate for the first time that pan-T cell, PD-1-mediated, exhaustion during VL influenced macrophage-reactive oxygen intermediate production. Blockade of the PD-1 pathway improved the ability of phagocytes isolated from dogs presenting with clinical VL to clear intracellular parasites. T cell exhaustion during symptomatic canine leishmaniasis has implications for the response to vaccination and therapeutic strategies for control of Leishmania infantum in this important reservoir species.
Collapse
Affiliation(s)
- Kevin J Esch
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA 50011
| | | | | | | | | |
Collapse
|