1
|
Mahor H, Mukherjee A, Sarkar A, Saha B. Anti-leishmanial therapy: Caught between drugs and immune targets. Exp Parasitol 2023; 245:108441. [PMID: 36572088 DOI: 10.1016/j.exppara.2022.108441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/12/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Leishmaniasis is an enigmatic disease that has very restricted options for chemotherapy and none for prophylaxis. As a result, deriving therapeutic principles for curing the disease has been a major objective in Leishmania research for a long time. Leishmania is a protozoan parasite that lives within macrophages by subverting or switching cell signaling to the pathways that ensure its intracellular survival. Therefore, three groups of molecules aimed at blocking or eliminating the parasite, at least, in principle, include blockers of macrophage receptor- Leishmania ligand interaction, macrophage-activating small molecules, peptides and cytokines, and signaling inhibitors or activators. Macrophages also act as an antigen-presenting cell, presenting antigen to the antigen-specific T cells to induce activation and differentiation of the effector T cell subsets that either execute or suppress anti-leishmanial functions. Three groups of therapeutic principles targeting this sphere of Leishmania-macrophage interaction include antibodies that block pro-leishmanial response of T cells, ligands that activate anti-leishmanial T cells and the antigens for therapeutic vaccines. Besides these, prophylactic vaccines have been in clinical trials but none has succeeded so far. Herein, we have attempted to encompass all these principles and compose a comprehensive review to analyze the feasibility and adoptability of different therapeutics for leishmaniasis.
Collapse
Affiliation(s)
- Hima Mahor
- National Centre for Cell Science, Ganeshkhind, Pune, 411007, India
| | - Arka Mukherjee
- Trident Academy of Creative Technology, Bhubaneswar, 751024, Odisha, India
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, 751024, Odisha, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune, 411007, India; Trident Academy of Creative Technology, Bhubaneswar, 751024, Odisha, India.
| |
Collapse
|
2
|
Gálvez NMS, Bohmwald K, Pacheco GA, Andrade CA, Carreño LJ, Kalergis AM. Type I Natural Killer T Cells as Key Regulators of the Immune Response to Infectious Diseases. Clin Microbiol Rev 2021; 34:e00232-20. [PMID: 33361143 PMCID: PMC7950362 DOI: 10.1128/cmr.00232-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The immune system must work in an orchestrated way to achieve an optimal response upon detection of antigens. The cells comprising the immune response are traditionally divided into two major subsets, innate and adaptive, with particular characteristics for each type. Type I natural killer T (iNKT) cells are defined as innate-like T cells sharing features with both traditional adaptive and innate cells, such as the expression of an invariant T cell receptor (TCR) and several NK receptors. The invariant TCR in iNKT cells interacts with CD1d, a major histocompatibility complex class I (MHC-I)-like molecule. CD1d can bind and present antigens of lipid nature and induce the activation of iNKT cells, leading to the secretion of various cytokines, such as gamma interferon (IFN-γ) and interleukin 4 (IL-4). These cytokines will aid in the activation of other immune cells following stimulation of iNKT cells. Several molecules with the capacity to bind to CD1d have been discovered, including α-galactosylceramide. Likewise, several molecules have been synthesized that are capable of polarizing iNKT cells into different profiles, either pro- or anti-inflammatory. This versatility allows NKT cells to either aid or impair the clearance of pathogens or to even control or increase the symptoms associated with pathogenic infections. Such diverse contributions of NKT cells to infectious diseases are supported by several publications showing either a beneficial or detrimental role of these cells during diseases. In this article, we discuss current data relative to iNKT cells and their features, with an emphasis on their driving role in diseases produced by pathogenic agents in an organ-oriented fashion.
Collapse
Affiliation(s)
- Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaspar A Pacheco
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leandro J Carreño
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
3
|
Cutaneous Leishmaniasis: The Complexity of Host's Effective Immune Response against a Polymorphic Parasitic Disease. J Immunol Res 2019; 2019:2603730. [PMID: 31871953 PMCID: PMC6913332 DOI: 10.1155/2019/2603730] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 10/05/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
Abstract
This review is aimed at providing a comprehensive outline of the immune response displayed against cutaneous leishmaniasis (CL), the more common zoonotic infection caused by protozoan parasites of the genus Leishmania. Although of polymorphic clinical presentation, classically CL is characterized by leishmaniotic lesions on the face and extremities of the patients, which can be ulcerative, and even after healing can lead to permanent injuries and disfigurement, affecting significantly their psychological, social, and economic well-being. According a report released by the World Health Organization, the disability-adjusted life years (DALYs) lost due to leishmaniasis are close to 2.4 million, annually there are 1.0–1.5 million new cases of CL, and a numerous population is at risk in the endemic areas. Despite its increasing worldwide incidence, it is one of the so-called neglected tropical diseases. Furthermore, this review provides an overview of the existing knowledge of the host innate and acquired immune response to cutaneous species of Leishmania. The use of animal models and of in vitro studies has improved the understanding of parasite-host interplay and the complexity of immune mechanisms involved. The importance of diagnosis accuracy associated with effective patient management in CL reduction is highlighted. However, the multiple factors involved in CL epizoology associated with the unavailability of vaccines or drugs to prevent infection make difficult to formulate an effective strategy for CL control.
Collapse
|
4
|
Galgamuwa LS, Sumanasena B, Iddawela D, Wickramasinghe S, Yatawara L. Assessment of intralesional cytokine profile of cutaneous leishmaniasis caused by Leishmania donovani in Sri Lanka. BMC Microbiol 2019; 19:14. [PMID: 30642262 PMCID: PMC6332851 DOI: 10.1186/s12866-018-1384-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 12/27/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cytokines play a vital role in the host immune response to infection by initiating the healing process and/or accelerating the progression of the disease in cutaneous leishmaniasis (CL). Very little evidence is available on cytokine profiles and their regulatory function in CL patients in Sri Lanka. The aim of this study was to determine the cytokine expression pattern of IFN-γ, IL-4, IL-11 and IL-12p40 in CL patients and in healthy volunteers. Patients with suspected CL lesions attending to the Dermatology Clinic at the Anuradhapura Teaching Hospital were included in the study. Reverse transcription real time polymerase chain reaction (real-time RT-PCR) was performed to determine the relative expression level of target cytokines. Expression levels were quantified by 2- ∆∆CT equation. RESULTS The expression of cytokines IFN-γ, IL-4, IL-11 and IL-12p40 were significantly higher in CL patients compared to healthy volunteers (p < 0.05). There was a significant association between the expression of IFN-γ and the duration of the lesion (p = 0.021). Wet CL lesions showed significantly higher expression of IL-4, IL-11 and IL-12p40 (p = 0.039, 0.018 and 0.021 respectively) compared to dry lesions. Papulo-nodular lesions showed significantly high expression of IFN-γ (p = 0.023). However, cytokine expression was not significantly associated with the number, size and the locations of lesions. CONCLUSIONS The expression levels of all cytokines tested in the present study were significantly (p < 0.05) high in CL patients. Th1 response (IFN-γ and IL-12p40) had higher expression levels compared to Th2 (IL-4) and IL-11 in CL patients.
Collapse
Affiliation(s)
| | - Buthsiri Sumanasena
- Anuradhapura Teaching Hospital, Harischandra Mawatha, Anuradhapura, Sri Lanka
| | - Devika Iddawela
- Department of Parasitology, Faculty of Medicine, University of Peradeniya, Peradeniya, 20400 Sri Lanka
| | - Susiji Wickramasinghe
- Department of Parasitology, Faculty of Medicine, University of Peradeniya, Peradeniya, 20400 Sri Lanka
| | - Lalani Yatawara
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, University of Peradeniya, Peradeniya, 20400 Sri Lanka
| |
Collapse
|
5
|
Synthetic analogs of an Entamoeba histolytica glycolipid designed to combat intracellular Leishmania infection. Sci Rep 2017; 7:9472. [PMID: 28842620 PMCID: PMC5572710 DOI: 10.1038/s41598-017-09894-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/31/2017] [Indexed: 11/19/2022] Open
Abstract
Intracellular pathogens belonging to the genus Leishmania have developed effective strategies that enable them to survive within host immune cells. Immunostimulatory compounds that counteract such immunological escape mechanisms represent promising treatment options for diseases. Here, we demonstrate that a lipopeptidephosphoglycan (LPPG) isolated from the membrane of a protozoan parasite, Entamoeba histolytica (Eh), shows considerable immunostimulatory effects targeted against Leishmania (L.) major, a representative species responsible for cutaneous leishmaniasis (CL). Treatment led to a marked reduction in the number of intracellular Leishmania parasites in vitro, and ameliorated CL in a mouse model. We next designed and synthesized analogs of the phosphatidylinositol anchors harbored by EhLPPG; two of these analogs reproduced the anti-leishmanial activity of the native compound by inducing production of pro-inflammatory cytokines. The use of such compounds, either alone or as a supportive option, might improve the currently unsatisfactory treatment of CL and other diseases caused by pathogen-manipulated immune responses.
Collapse
|
6
|
Zamora-Chimal J, Hernández-Ruiz J, Becker I. NKT cells in leishmaniasis. Immunobiology 2017; 222:641-646. [DOI: 10.1016/j.imbio.2016.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 11/30/2016] [Indexed: 10/20/2022]
|
7
|
Abstract
Diseases caused by Leishmania present a worldwide problem, and current therapeutic approaches are unable to achieve a sterile cure. Leishmania is able to persist in host cells by evading or exploiting host immune mechanisms. A thorough understanding of these mechanisms could lead to better strategies for effective management of Leishmania infections. Current research has focused on parasite modification of host cell signaling pathways, entry into phagocytic cells, and modulation of cytokine and chemokine profiles that alter immune cell activation and trafficking to sites of infection. Immuno-therapeutic approaches that target these mechanisms of immune evasion by Leishmania offer promising areas for preclinical and clinical research.
Collapse
|
8
|
Cecílio P, Pérez-Cabezas B, Santarém N, Maciel J, Rodrigues V, Cordeiro da Silva A. Deception and manipulation: the arms of leishmania, a successful parasite. Front Immunol 2014; 5:480. [PMID: 25368612 PMCID: PMC4202772 DOI: 10.3389/fimmu.2014.00480] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/19/2014] [Indexed: 12/12/2022] Open
Abstract
Leishmania spp. are intracellular parasitic protozoa responsible for a group of neglected tropical diseases, endemic in 98 countries around the world, called leishmaniasis. These parasites have a complex digenetic life cycle requiring a susceptible vertebrate host and a permissive insect vector, which allow their transmission. The clinical manifestations associated with leishmaniasis depend on complex interactions between the parasite and the host immune system. Consequently, leishmaniasis can be manifested as a self-healing cutaneous affliction or a visceral pathology, being the last one fatal in 85–90% of untreated cases. As a result of a long host–parasite co-evolutionary process, Leishmania spp. developed different immunomodulatory strategies that are essential for the establishment of infection. Only through deception and manipulation of the immune system, Leishmania spp. can complete its life cycle and survive. The understanding of the mechanisms associated with immune evasion and disease progression is essential for the development of novel therapies and vaccine approaches. Here, we revise how the parasite manipulates cell death and immune responses to survive and thrive in the shadow of the immune system.
Collapse
Affiliation(s)
- Pedro Cecílio
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Begoña Pérez-Cabezas
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Nuno Santarém
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Joana Maciel
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Vasco Rodrigues
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal
| | - Anabela Cordeiro da Silva
- Parasite Disease Group, Institute for Molecular and Cell Biology (IBMC), University of Porto , Porto , Portugal ; Department of Biological Sciences, Faculty of Pharmacy, University of Porto , Porto , Portugal
| |
Collapse
|
9
|
Griewank KG, Lorenz B, Fischer MR, Boon L, Lopez Kostka S, von Stebut E. Immune modulating effects of NKT cells in a physiologically low dose Leishmania major infection model after αGalCer analog PBS57 stimulation. PLoS Negl Trop Dis 2014; 8:e2917. [PMID: 24967701 PMCID: PMC4072590 DOI: 10.1371/journal.pntd.0002917] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/17/2014] [Indexed: 11/18/2022] Open
Abstract
Leishmaniasis is a parasitic infection affecting ∼12 million people worldwide, mostly in developing countries. Treatment options are limited and no effective vaccines exist to date. Natural Killer T (NKT) cells are a conserved innate-like lymphocyte population with immunomodulating effects in various settings. A number of reports state a role of NKT cells in different models of Leishmania infection. Here, we investigated the effect of NKT cells in a physiologically relevant, intradermal low dose infection model. After inoculation of 103 infectious-stage L. major, comparable numbers of skin-immigrating NKT cells in both susceptible BALB/c mice and resistant C57BL/6 mice were noted. Compared to their wild type counterparts, NKT cell-deficient mice on a C57BL/6 background were better able to contain infection with L. major and showed decreased IL-4 production in cytokine analysis performed 5 and 8 weeks after infection. Low doses of the NKT cell stimulating αGalCer analog PBS57 applied at the time of infection led to disease exacerbation in C57BL/6 wild-type, but not NKT-deficient mice. The effect was dependent both on the timing and amount of PBS57 administered. The effect of NKT cell stimulation by PBS57 proved to be IL-4 dependent, as it was neutralized in IL-4-deficient C57BL/6 or anti-IL-4 antibody-treated wild-type mice. In contrast to C57BL/6 mice, administration of PBS57 in susceptible BALB/c mice resulted in an improved course of disease. Our results reveal a strain- and cytokine-dependent regulatory role of NKT cells in the development of immunity to low dose L. major infections. These effects, probably masked in previous studies using higher parasite inocula, should be considered in future therapy and immunization approaches. Cutaneous leishmaniasis is a disease affecting about 12 million people worldwide. It is transmitted by a sand fly and primarily affects people in developing countries. To date there are no effective vaccines. Many of the treatments available have serious side effects and resistance mechanisms are becoming an increasingly prevalent problem. Natural killer T (NKT) cells are a unique T cell population recognizing glycolipids. Their role in immune processes, especially in infectious diseases, is incompletely understood. In the current study, we investigated the role of NKT cells in Leishmania infections in detail. We found that NKT cells can significantly alter the development of immunity, however in different directions depending on the host's genetic background. Their natural effect on infection can be increased when applying the stimulating antigen alpha-Galactosyl-Ceramide (αGalCer) or its analogs (in our study PBS57). Our results show that the effect of these cells in resistant mice (which are generally reminiscent of the situation in humans) is largely mediated by cytokine secretion, in particular IL-4, a Th2 cytokine. We conclude that NKT cells influence the course of Leishmania infection and that therapeutically modulating their function could be beneficial both to treat existing infections, as well as potentially develop desperately needed, effective vaccines.
Collapse
Affiliation(s)
- Klaus G. Griewank
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- Department of Dermatology, University Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Beate Lorenz
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Michael R. Fischer
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | | | - Susanna Lopez Kostka
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Esther von Stebut
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- * E-mail:
| |
Collapse
|
10
|
Kinjo Y, Kitano N, Kronenberg M. The role of invariant natural killer T cells in microbial immunity. J Infect Chemother 2013; 19:560-70. [PMID: 23846426 PMCID: PMC3822041 DOI: 10.1007/s10156-013-0638-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Indexed: 10/26/2022]
Abstract
Invariant natural killer T cells (iNKT cells) are unique lymphocytes with characteristic features, such as expression of an invariant T-cell antigen receptor (TCR) α-chain, recognition of glycolipid antigens presented by CD1d molecules, and ability to rapidly produce large amounts of cytokines, including interferon-γ (IFN-γ) and interleukin 4 (IL-4) upon TCR stimulation. Many studies have demonstrated that iNKT cells participate in immune response against diverse microbes, including bacteria, fungi, protozoan parasites, and viruses. Generally, these cells play protective roles in host defense against infections. However, in some contexts they play pathogenic roles, by inducing or augmenting inflammation. Recent reports show that iNKT cells recognize glycolipid antigens from pathogenic bacteria including Streptococcus pneumoniae, and they contribute to host defense against infection. iNKT cell responses to these microbial glycolipid antigens are highly conserved between rodents and humans, suggesting that iNKT cells are evolutionally conserved because their invariant TCR is useful in detecting certain pathogens. Furthermore, glycolipid-mediated iNKT cell activation during immunization has adjuvant activity, enhancing humoral and cell-mediated responses. Therefore, iNKT cell activation is an attractive target for developing new vaccines for infectious diseases.
Collapse
Affiliation(s)
- Yuki Kinjo
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo, 162-8640, Japan.
| | | | | |
Collapse
|
11
|
Bogdan C. Natural killer cells in experimental and human leishmaniasis. Front Cell Infect Microbiol 2012; 2:69. [PMID: 22919660 PMCID: PMC3417408 DOI: 10.3389/fcimb.2012.00069] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Accepted: 05/02/2012] [Indexed: 12/27/2022] Open
Abstract
Infections with parasites of the genus Leishmania lead to a rapid, but transient activation of natural killer (NK) cells. In mice activation of NK cells requires a toll-like-receptor 9-dependent stimulation of dendritic cells (DC) which is followed by the production of IL-12. Although NK cells appear to be non-essential for the ultimate control of cutaneous and visceral leishmaniasis (VL) and can exhibit immunosuppressive functions, they form an important source of interferon (IFN)-γ, which elicits antileishmanial activity in macrophages and helps to pave a protective T helper cell response. In contrast, the cytotoxic activity of NK cells is dispensable, because Leishmania-infected myeloid cells are largely resistant to NK-mediated lysis. In human cutaneous and VL, the functional importance of NK cells is suggested by reports that demonstrate (1) a direct activation or inhibition of NK cells by Leishmania promastigotes, (2) the suppression of NK cell numbers or activity during chronic, non-healing infections, and (3) the recovery of NK cell activity following treatment. This review aims to provide an integrated view on the migration, activation, inhibition, function, and therapeutic modulation of NK cells in experimental and human leishmaniasis.
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen Bavaria, Germany. christian.bogdan@ uk-erlangen.de
| |
Collapse
|
12
|
Invariant NKT cells drive hepatic cytokinic microenvironment favoring efficient granuloma formation and early control of Leishmania donovani infection. PLoS One 2012; 7:e33413. [PMID: 22457760 PMCID: PMC3310876 DOI: 10.1371/journal.pone.0033413] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 02/08/2012] [Indexed: 12/22/2022] Open
Abstract
The development of inflammatory granulomas around infected Kupffer cells is necessary for hepatic parasite clearance during visceral leishmaniasis. Invariant NKT (iNKT) cells are predominant T cells in the mouse liver and can synthesize large quantities of IL-4 and IFN-γ, two cytokines involved in granuloma formation. This study analyzed the role of iNKT cells in the hepatic immune response during Leishmania donovani infection, using a murine model of wild-type (WT) and iNKT cell-deficient (Jα18-/-) C57BL/6 mice sacrificed 15, 30 or 60 days post-infection. We recorded hepatic parasite loads, cytokine expression, and analyzed granulomatous response by immunohistochemistry and hepatic immune cell infiltration by flow cytometry. Whereas WT animals rapidly controlled the infection and developed an inflammatory response associated with a massive influx of iNKT cells observed by flow cytometry, Jα18-/- mice had significantly higher parasitic loads on all time points. This lack of control of parasite burden was associated with a delay in granuloma maturation (28.1% of large granulomas at day 60 versus 50.7% in WT). Cytokine transcriptome analysis showed that mRNA of 90/101 genes encoding chemokines, cytokines and their receptors, was underexpressed in Jα18-/- mice. Detection of IL-4 and TNF-α by ELISA in liver extracts was also significantly lower in Jα18-/- mice. Consistent with flow cytometry analysis, cytokinome profile in WT mice showed a bias of expression towards T cell-chemoattractant chemokines on D15, and displayed a switch towards expression of granulocytes and/or monocytes -chemoattractant chemokines on D60. In Jα18-/- mice, the significantly lower expression of CXCL5, MIP-2 and CCL2 mRNA was correlated with a defect in myeloperoxidase positive-cell attraction observed by immunohistochemistry and with a lower granulocyte and monocyte infiltration in the liver, as shown by flow cytometry. These data indicate that iNKT cells play a role in early and sustained pro-inflammatory cytokine response warranting efficient organization of hepatic granulomas and parasite clearance.
Collapse
|
13
|
Duwaerts CC, Gregory SH. Targeting the diverse immunological functions expressed by hepatic NKT cells. Expert Opin Ther Targets 2011; 15:973-88. [PMID: 21564001 DOI: 10.1517/14728222.2011.584874] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION NKT cells comprise approximately 30% of the hepatic lymphoid population in mice (∼ 50% in humans). Most mouse hepatic NKT cells [invariant (i)NKT cells] express T cell receptors, composed of invariant Vα14Jα18 chains. Unlike conventional T cells, iNKT cells recognize glycolipids presented in association with MHC class Ib (CD1d) molecules. Purportedly, iNKT cells serve key functions in several immunological events; the nature of these is often unclear. The consequences of hepatic iNKT cell activation can be beneficial or detrimental. α-Galactosylceramide stimulates the production of IFN-γ and IL-4. The reciprocal suppression exhibited by these cytokines limits the potential therapeutic value of α-galactosylceramide. Efforts are ongoing to develop α-galactosylceramide analogs that modulate iNKT cell activity and selectively promote IFN-γ or IL-4. AREAS COVERED An overview of hepatic iNKT cells and their purported role in liver disease. Efforts to develop therapeutic agents that promote their beneficial contributions. EXPERT OPINION While a growing body of literature documents the differential effects of α-GalCer analogs on IFN-γ and IL-4 production, the effects of these analogs on other iNKT cell activities remain to be determined. An exhaustive examination of the effects of these analogs on inflammation and liver injury in animal models remains prior to considering their utility in clinical trials.
Collapse
Affiliation(s)
- Caroline C Duwaerts
- Rhode Island Hospital and The Warren Alpert Medical School at Brown University, Department of Medicine, Providence, RI 02903, USA
| | | |
Collapse
|
14
|
Zeissig S, Dougan SK, Barral DC, Junker Y, Chen Z, Kaser A, Ho M, Mandel H, McIntyre A, Kennedy SM, Painter GF, Veerapen N, Besra GS, Cerundolo V, Yue S, Beladi S, Behar SM, Chen X, Gumperz JE, Breckpot K, Raper A, Baer A, Exley MA, Hegele RA, Cuchel M, Rader DJ, Davidson NO, Blumberg RS. Primary deficiency of microsomal triglyceride transfer protein in human abetalipoproteinemia is associated with loss of CD1 function. J Clin Invest 2010; 120:2889-99. [PMID: 20592474 DOI: 10.1172/jci42703] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 05/10/2010] [Indexed: 01/07/2023] Open
Abstract
Abetalipoproteinemia (ABL) is a rare Mendelian disorder of lipid metabolism due to genetic deficiency in microsomal triglyceride transfer protein (MTP). It is associated with defects in MTP-mediated lipid transfer onto apolipoprotein B (APOB) and impaired secretion of APOB-containing lipoproteins. Recently, MTP was shown to regulate the CD1 family of lipid antigen-presenting molecules, but little is known about immune function in ABL patients. Here, we have shown that ABL is characterized by immune defects affecting presentation of self and microbial lipid antigens by group 1 (CD1a, CD1b, CD1c) and group 2 (CD1d) CD1 molecules. In dendritic cells isolated from ABL patients, MTP deficiency was associated with increased proteasomal degradation of group 1 CD1 molecules. Although CD1d escaped degradation, it was unable to load antigens and exhibited functional defects similar to those affecting the group 1 CD1 molecules. The reduction in CD1 function resulted in impaired activation of CD1-restricted T and invariant natural killer T (iNKT) cells and reduced numbers and phenotypic alterations of iNKT cells consistent with central and peripheral CD1 defects in vivo. These data highlight MTP as a unique regulator of human metabolic and immune pathways and reveal that ABL is not only a disorder of lipid metabolism but also an immune disease involving CD1.
Collapse
Affiliation(s)
- Sebastian Zeissig
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Gan LH, Pan YQ, Xu DP, Li M, Lin A, Yan WH. Polymorphism of human CD1a, CD1d, and CD1e in exon 2 in Chinese Han and She ethnic populations. ACTA ACUST UNITED AC 2010; 75:691-5. [DOI: 10.1111/j.1399-0039.2010.01443.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
Vaccination with live Leishmania major and CpG DNA promotes interleukin-2 production by dermal dendritic cells and NK cell activation. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:1601-6. [PMID: 19776191 DOI: 10.1128/cvi.00249-09] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cutaneous leishmaniasis due to Leishmania major is an emerging, chronic parasitic disease that causes disfigurement and social stigmatization. Drug therapy is inadequate, and there is no vaccine. Inoculation of virulent parasites (leishmanization) is the only intervention that has ever provided protection, because it mimics natural infection and immunity, but it was discontinued due to safety concerns (uncontrolled vaccinal lesions). In an effort to retain the benefits (immunity) while avoiding the side effects (lesions) of leishmanization, we immunized C57BL/6 mice with L. major and CpG DNA (Lm/CpG). This combination prevented lesions while inducing immunity. Also, the vaccination with live parasites and the Toll-like receptor 9 agonist enhanced innate immune responses by activating dermal dendritic cells (DCs) to produce cytokines. Here we report that the Lm/CpG vaccine induced dermal DCs, but not bone marrow-derived DCs, to produce interleukin-2 (IL-2). The release of this unusual DC-derived cytokine was concomitant with a peak in numbers of NK cells that produced gamma interferon (IFN-gamma) and also enhanced activation of proliferation of IFN-gamma+ CD4+ T cells. Parasite growth was controlled in Lm/CpG-vaccinated animals. This is the first demonstration of the ability of dermal DCs to produce IL-2 and of the activation of NK cells by vaccination in the context of leishmaniasis. Understanding how the Lm/CpG vaccine enhances innate immunity may provide new tools to develop vaccines against L. major, other chronic infectious diseases, or other conditions, such as cancer.
Collapse
|
17
|
Tessmer MS, Fatima A, Paget C, Trottein F, Brossay L. NKT cell immune responses to viral infection. Expert Opin Ther Targets 2009; 13:153-62. [PMID: 19236234 DOI: 10.1517/14712590802653601] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Natural killer T (NKT) cells are a heterogeneous population of innate T cells that have attracted interest because of their potential to regulate immune responses to a variety of pathogens. The most widely studied NKT cell subset is the invariant (i)NKT cells that recognize glycolipids in the context of the CD1d molecule. The multifaceted methods of activation iNKT cells possess and their ability to produce regulatory cytokines has made them a primary target for studies. OBJECTIVE/METHODS To give insights into the roles of iNKT cells during infectious diseases, particularly viral infections. We also highlight mechanisms leading to iNKT cell activation in response to pathogens. CONCLUSIONS iNKT cell's versatility allows them to detect and respond to several viruses. Therapeutic approaches to specifically target iNKT cells will require additional research. Notably, the roles of non-invariant NKT cells in response to pathogens warrant further investigation.
Collapse
Affiliation(s)
- Marlowe S Tessmer
- Brown University, Department of Molecular Microbiology and Immunology, Providence, USA
| | | | | | | | | |
Collapse
|
18
|
|
19
|
Kinjo Y, Kronenberg M. Detection of microbes by natural killer T cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 633:17-26. [PMID: 19209678 DOI: 10.1007/978-0-387-79311-5_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Yuki Kinjo
- La Jolla Institute for Allergy and Immunology, CA 92037, USA
| | | |
Collapse
|
20
|
Ojeda SS, Wang ZJ, Mares CA, Chang TA, Li Q, Morris EG, Jerabek PA, Teale JM. Rapid dissemination of Francisella tularensis and the effect of route of infection. BMC Microbiol 2008; 8:215. [PMID: 19068128 PMCID: PMC2651876 DOI: 10.1186/1471-2180-8-215] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Accepted: 12/09/2008] [Indexed: 12/14/2022] Open
Abstract
Background Francisella tularensis subsp. tularensis is classified as a Category A bioweapon that is capable of establishing a lethal infection in humans upon inhalation of very few organisms. However, the virulence mechanisms of this organism are not well characterized. Francisella tularensis subsp. novicida, which is an equally virulent subspecies in mice, was used in concert with a microPET scanner to better understand its temporal dissemination in vivo upon intranasal infection and how such dissemination compares with other routes of infection. Adult mice were inoculated intranasally with F. tularensis subsp. novicida radiolabeled with 64Cu and imaged by microPET at 0.25, 2 and 20 hours post-infection. Results 64Cu labeled F. tularensis subsp. novicida administered intranasally or intratracheally were visualized in the respiratory tract and stomach at 0.25 hours post infection. By 20 hours, there was significant tropism to the lung compared with other tissues. In contrast, the images of radiolabeled F. tularensis subsp. novicida when administered intragastrically, intradermally, intraperitoneally and intravenouslly were more generally limited to the gastrointestinal system, site of inoculation, liver and spleen respectively. MicroPET images correlated with the biodistribution of isotope and bacterial burdens in analyzed tissues. Conclusion Our findings suggest that Francisella has a differential tissue tropism depending on the route of entry and that the virulence of Francisella by the pulmonary route is associated with a rapid bacteremia and an early preferential tropism to the lung. In addition, the use of the microPET device allowed us to identify the cecum as a novel site of colonization of Francisella tularensis subsp. novicida in mice.
Collapse
Affiliation(s)
- Sandra S Ojeda
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Dondji B, Deak E, Goldsmith-Pestana K, Perez-Jimenez E, Esteban M, Miyake S, Yamamura T, McMahon-Pratt D. Intradermal NKT cell activation during DNA priming in heterologous prime-boost vaccination enhances T cell responses and protection against Leishmania. Eur J Immunol 2008; 38:706-19. [PMID: 18286565 DOI: 10.1002/eji.200737660] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Heterologous prime-boost vaccination employing DNA-vaccinia virus (VACV) modality using the Leishmania homologue of receptors for activated C kinase (LACK) (p36) antigen has been shown to elicit protective immunity against both murine cutaneous and visceral leishmaniasis. However, DNA priming is known to have limited efficacy; therefore in the current study the effect of NKT cell activation using alpha-galactosyl-ceramide (alphaGalCer) during intradermal DNAp36 priming was examined. Vaccinated mice receiving alphaGalCer + DNAp36 followed by a boost with VVp36 appeared to be resolving their lesions and had at ten- to 20-fold higher reductions in parasite burdens. NKT cell activation during alphaGalCer + DNAp36 priming resulted in higher numbers of antigen-reactive effector CD4(+) and CD8(+) T cells producing granzyme and IFN-gamma, with lower levels of IL-10. Although immunodepletion studies indicate that both CD4 and CD8 T cells provide protection in the vaccinated mice, the contribution of CD4(+) T cells was significantly increased in mice primed with DNAp36 together with alphaGalCer. Notably 5 months after boosting, mice vaccinated with DNAp36 + alphaGalCer continued to show sustained and heightened T cell immune responses. Thus, heterologous prime-boost vaccination using alphaGalCer during priming is highly protective against murine cutaneous leishmaniasis, resulting in the heightened activation and development of CD4 and CD8 T cells (effector and memory T cells).
Collapse
Affiliation(s)
- Blaise Dondji
- Department of Epidemiology & Public Health, Yale University School of Medicine, New Haven, CT 06520-8034, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Wiethe C, Debus A, Mohrs M, Steinkasserer A, Lutz M, Gessner A. Dendritic Cell Differentiation State and Their Interaction with NKT Cells Determine Th1/Th2 Differentiation in the Murine Model ofLeishmania majorInfection. THE JOURNAL OF IMMUNOLOGY 2008; 180:4371-81. [DOI: 10.4049/jimmunol.180.7.4371] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
23
|
Stanley AC, Zhou Y, Amante FH, Randall LM, Haque A, Pellicci DG, Hill GR, Smyth MJ, Godfrey DI, Engwerda CR. Activation of invariant NKT cells exacerbates experimental visceral leishmaniasis. PLoS Pathog 2008; 4:e1000028. [PMID: 18463695 PMCID: PMC2265425 DOI: 10.1371/journal.ppat.1000028] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 01/07/2008] [Indexed: 11/22/2022] Open
Abstract
We report that natural killer T (NKT) cells play only a minor physiological role in protection from Leishmania donovani infection in C57BL/6 mice. Furthermore, attempts at therapeutic activation of invariant NKT (iNKT) cells with α-galactosylceramide (α-GalCer) during L. donovani infection exacerbated, rather than ameliorated, experimental visceral leishmaniasis. The inability of α-GalCer to promote anti-parasitic immunity did not result from inefficient antigen presentation caused by infection because α-GalCer–loaded bone marrow–derived dendritic cells were also unable to improve disease resolution. The immune-dampening affect of α-GalCer correlated with a bias towards increased IL-4 production by iNKT cells following α-GalCer stimulation in infected mice compared to naïve controls. However, studies in IL-4–deficient mice, and IL-4 neutralisation in cytokine-sufficient mice revealed that α-GalCer–induced IL-4 production during infection had only a minor role in impaired parasite control. Analysis of liver cell composition following α-GalCer stimulation during an established L. donovani infection revealed important differences, predominantly a decrease in IFNγ+ CD8+ T cells, compared with control-treated mice. Our data clearly illustrate the double-edged sword of NKT cell–based therapy, showing that in some circumstances, such as when sub-clinical or chronic infections exist, iNKT cell activation can have adverse outcomes. Natural killer T (NKT) cells are a unique subset of T cells that can produce large quantities of inflammatory cytokines very rapidly upon stimulation. They are known to be strongly stimulated by a molecule called α-galactosylceramide (α-GalCer) that is derived from a marine sponge, and in this way α-GalCer is hoped to provide effective immunotherapy for a wide range of diseases. We attempted to stimulate NKT cells with α-GalCer in mice infected with Leishmania donovani, a protozoan parasite that causes a chronic disease known as visceral leishmaniasis in humans. L. donovani characteristically causes an acute resolving infection in the liver where NKT cells are abundant. Therefore, we hypothesised that by stimulating these cells with α-GalCer we would improve the rate of hepatic disease resolution. However, while α-GalCer administered prior to infection had no effect on hepatic parasite burden, α-GalCer administered during an established infection exacerbated hepatic disease, associated with a decrease in IFNγ-producing CD8+ T cells. These results are important as they demonstrate that therapies aimed at modulating NKT cell function are not always beneficial, and adverse consequences may occur in certain circumstances, such as in the presence of persistent and/or sub-clinical infections.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/pharmacology
- Biomarkers
- CD8-Positive T-Lymphocytes
- Disease Models, Animal
- Female
- Galactosylceramides/therapeutic use
- Gene Silencing
- Host-Parasite Interactions
- Immunologic Factors/therapeutic use
- Interferon-gamma
- Interleukin-4/deficiency
- Interleukin-4/immunology
- Interleukin-4/metabolism
- Killer Cells, Natural/immunology
- Leishmania donovani/immunology
- Leishmania donovani/pathogenicity
- Leishmaniasis, Visceral/drug therapy
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/pathology
- Liver/drug effects
- Liver/metabolism
- Liver/parasitology
- Lymphocyte Activation
- Lymphocyte Subsets/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Amanda C. Stanley
- Immunology and Infection Laboratory, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Yonghong Zhou
- Immunology and Infection Laboratory, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Fiona H. Amante
- Immunology and Infection Laboratory, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Louise M. Randall
- Immunology and Infection Laboratory, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Ashraful Haque
- Immunology and Infection Laboratory, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Daniel G. Pellicci
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | - Geoff R. Hill
- Immunology and Infection Laboratory, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Mark J. Smyth
- Cancer Immunology Program, Trescowthick Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | - Christian R. Engwerda
- Immunology and Infection Laboratory, Queensland Institute of Medical Research, Herston, Queensland, Australia
- * E-mail:
| |
Collapse
|
24
|
Silvestre R, Cordeiro-Da-Silva A, Santarém N, Vergnes B, Sereno D, Ouaissi A. SIR2-deficient Leishmania infantum induces a defined IFN-gamma/IL-10 pattern that correlates with protection. THE JOURNAL OF IMMUNOLOGY 2007; 179:3161-70. [PMID: 17709531 DOI: 10.4049/jimmunol.179.5.3161] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ability to manipulate the Leishmania genome to create genetically modified parasites by introducing or eliminating genes is considered a powerful alternative for developing a new generation vaccine against leishmaniasis. Previously, we showed that the deletion of one allele of the Leishmania infantum silent information regulatory 2 (LiSIR2) locus was sufficient to dramatically affect amastigote axenic proliferation. Furthermore, LiSIR2 single knockout (LiSIR2(+/-)) amastigotes were unable to replicate in vitro inside macrophages. Because this L. infantum mutant persisted in BALB/c mice for up to 6 wk but failed to establish an infection, we tested its ability to provide protection toward a virulent L. infantum challenge. Strikingly, vaccination with a single i.p. injection of LiSIR2(+/-) single knockout elicits complete protection. Thus, vaccinated BALB/c mice showed a reversal of T cell anergy with specific anti-Leishmania cytotoxic activity and high levels of NO production. Moreover, vaccinated mice simultaneously generated specific anti-Leishmania IgG Ab subclasses suggestive of both type 1 and type 2 responses. A strong correlation was found between the elimination of the parasites and an increased Leishmania-specific IFN-gamma/IL-10 ratio. Therefore, we propose that the polarization to a high IFN-gamma/low IL-10 ratio after challenge is a clear indicator of vaccine success. Furthermore these mutants, which presented attenuated virulence, represent a good model to understand the correlatives of protection in visceral leishmaniasis.
Collapse
Affiliation(s)
- Ricardo Silvestre
- Departamento de Bioquímica da Faculdade de Farmácia da Universidade do Porto, Portugal
| | | | | | | | | | | |
Collapse
|
25
|
Donovan MJ, Jayakumar A, McDowell MA. Inhibition of groups 1 and 2 CD1 molecules on human dendritic cells by Leishmania species. Parasite Immunol 2007; 29:515-24. [PMID: 17883454 DOI: 10.1111/j.1365-3024.2007.00970.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dendritic cells are potent immune-activating cells, which traditionally are thought of as presenters of protein antigen to lymphocytes to initiate an immune response. Recently, another mechanism of immune surveillance, the detection of lipid antigens, has been found to be mediated by the nonpolymorphic family of CD1 molecules. There are two different CD1 families, Group 1 consisting of CD1a, CD1b and CD1c, and Group 2 consisting only of CD1d. Leishmania donovani-infected dendritic cells have previously been demonstrated to exhibit decreased surface levels of Group 1 CD1 molecules and are no longer able to initiate a CD1b-restricted T cell response. In contrast to L. donovani, which disseminates to the visceral organs, L. major remains localized, forming a cutaneous lesion. We investigate here whether L. major, the aetiological agent of cutaneous leishmaniasis, also inhibits CD1 expression. We demonstrate that infection of human monocyte-derived dendritic cells with either L. major or L. donovani results in transcriptional down-regulation of both Groups 1 and 2 CD1 molecules. Furthermore, infection of monocytes during differentiation results in a cell phenotype similar to undifferentiated monocytes. Finally, we demonstrate that this down-regulation is not mediated by lipophosphoglycan or other phosphoglycans.
Collapse
Affiliation(s)
- M J Donovan
- Center for Global Health and Infectious Diseases, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46656, USA
| | | | | |
Collapse
|
26
|
Behar SM, Porcelli SA. CD1-restricted T cells in host defense to infectious diseases. Curr Top Microbiol Immunol 2007; 314:215-50. [PMID: 17593663 DOI: 10.1007/978-3-540-69511-0_9] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CD1 has been clearly shown to function as a microbial recognition system for activation of T cell responses, but its importance for mammalian protective responses against infections is still uncertain. The function of the group 1 CD1 isoforms, including human CD1a, CDlb, and CDLc, seems closely linked to adaptive immunity. These CD1 molecules control the responses of T cells that are highly specific for particular lipid antigens, the best known of which are abundantly expressed by pathogenic mycobacteria such as Mycobacterium tuberculosis and Mycobacterium leprae. Studies done mainly on human circulating T cells ex vivo support a significant role for group I CD1-restricted T cells in protective immunity to mycobacteria and potentially other pathogens, although supportive data from animal models is currently limited. In contrast, group 2 CD1 molecules, which include human CD1d and its orthologs, have been predominantly associated with the activation of CD1d-restricted NKT cells, which appear to be more appropriately viewed as a facet of the innate immune system. Whereas the recognition of certain self-lipid ligands by CD d-restricted NKT cells is well accepted, the importance of these T cells in mediating adaptive immune recognition of specific microbial lipid antigens remains controversial. Despite continuing uncertainty about the role of CD 1d-restricted NKT cells in natural infections, studies in mouse models demonstrate the potential of these T cells to exert various effects on a wide spectrum of infectious diseases, most likely by serving as a bridge between innate and adaptive immune responses.
Collapse
Affiliation(s)
- S M Behar
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Smith Building Room 518, One Jimmy Fund Way, Boston, MA 02115, USA
| | | |
Collapse
|
27
|
Tupin E, Kinjo Y, Kronenberg M. The unique role of natural killer T cells in the response to microorganisms. Nat Rev Microbiol 2007; 5:405-17. [PMID: 17487145 DOI: 10.1038/nrmicro1657] [Citation(s) in RCA: 355] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Natural killer T (NKT) cells combine features of the innate and adaptive immune systems. Recently, it has become evident that these T cells have crucial roles in the response to infectious agents. The antigen receptor expressed by NKT cells directly recognizes unusual glycolipids that are part of the membrane of certain Gram-negative bacteria and spirochetes. Moreover, even in the absence of microbial glycolipid antigens, these T cells respond to innate cytokines produced by dendritic cells that have been activated by microbes. This indirect sensing of infection, by responding to cytokines from activated dendritic cells, allows NKT cells to react to a broad range of infectious agents.
Collapse
Affiliation(s)
- Emmanuel Tupin
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | | | | |
Collapse
|