1
|
Osero BO, Aruleba RT, Brombacher F, Hurdayal R. Unravelling the unsolved paradoxes of cytokine families in host resistance and susceptibility to Leishmania infection. Cytokine X 2020; 2:100043. [PMID: 33415318 PMCID: PMC7773805 DOI: 10.1016/j.cytox.2020.100043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/16/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022] Open
Abstract
Leishmaniasis is a neglected disease caused by protozoan parasites of the genus Leishmania. Successful clearance of Leishmania relies on a robust human immune response and various cytokines have been implicated in resistance and susceptibility to Leishmania infection. Accordingly, various immunotherapeutic approaches involving cytokines and cytokine receptors are being considered as novel avenues of treatment given the limited efficacy of current anti-leishmanial drugs. These approaches target canonical T helper (Th)1/Type 1 cytokines as intended mediators of host-protection to infection whilst concomitantly suppressing Th2/Type 2 cytokines and their anticipated disease-promoting roles. However, the use of cytokine and cytokine receptor gene-deficient mice over the years has challenged this simplistic view of Th1/Type 1-mediated resistance and Th2/Type 2-mediated susceptibility. Indeed, contribution to susceptibility vs resistance is only a partial consequence to cytokine action as the overall response is multi-faceted due to the pleiotropic, redundant, antagonistic and synergistic action of cytokines and interactions with immune cells in the diseased state. Notably, while the responses of certain cytokines are selectively host-protective or characteristic disease-enhancers, some ligands exert a response depending on the parasite-species initiating infection. Paradoxically, others play dual or contradictory roles in different Leishmania immunopathologies. Hence, cytokines in disease is an unsolved paradox and a comprehensive knowledge of cytokine interplay is important to guide the development of novel immunotherapeutics against leishmaniasis. In this review, we characterize various cytokine families in persistence and clearance of the Leishmania parasite and particularly elucidate unsolved cytokine puzzles in leishmaniasis based on information acquired from "gain of knowledge by loss of function" studies in cytokine and cytokine receptor gene-deficient mice.
Collapse
Affiliation(s)
- Bernard Ong'ondo Osero
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Raphael Taiwo Aruleba
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Ramona Hurdayal
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| |
Collapse
|
2
|
Vacas A, Fernández-Rubio C, Larrea E, Peña-Guerrero J, Nguewa PA. LmjF.22.0810 from Leishmania major Modulates the Th2-Type Immune Response and Is Involved in Leishmaniasis Outcome. Biomedicines 2020; 8:biomedicines8110452. [PMID: 33114674 PMCID: PMC7692454 DOI: 10.3390/biomedicines8110452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 11/16/2022] Open
Abstract
A novel serine/threonine protein kinase, LmjF.22.0810, was recently described in Leishmania major. After generating an L. major cell line overexpressing LmjF.22.0810 (named LmJ3OE), the ability of this novel protein to modulate the Th2-type immune response was analyzed. Our results suggest that the protein kinase LmjF.22.0810 might be involved in leishmaniasis outcomes. Indeed, our study outlined the LmJ3OE parasites infectivity in vitro and in vivo. Transgenic parasites displayed lower phagocytosis rates in vitro, and their promastigote forms exhibited lower expression levels of virulence factors compared to their counterparts in control parasites. In addition, LmJ3OE parasites developed significantly smaller footpad swelling in susceptible BALB/c mice. Hematoxylin-eosin staining allowed the observation of a lower inflammatory infiltrate in the footpad from LmJ3OE-infected mice compared to animals inoculated with control parasites. Gene expression of Th2-associated cytokines and effectors revealed a dramatically lower induction in interleukin (IL)-4, IL-10, and arginase 1 (ARG1) mRNA levels at the beginning of the swelling; no expression change was found in Th1-associated cytokines except for IL-12. Accordingly, such results were validated by immunohistochemistry studies, illustrating a weaker expression of ARG1 and a similar induction for inducible NO synthase (iNOS) in footpads from LmJ3OE-infected mice compared to control L. major infected animals. Furthermore, the parasite burden was lower in footpads from LmJ3OE-infected mice. Our analysis indicated that such significant smaller footpad swellings might be due to an impairment of the Th2 immune response that subsequently benefits Th1 prevalence. Altogether, these studies depict LmjF.22.0810 as a potential modulator of host immune responses to Leishmania. Finally, this promising target might be involved in the modulation of infection outcome.
Collapse
Affiliation(s)
- Andrés Vacas
- Department of Microbiology and Parasitology, ISTUN Institute of Tropical Health, IdiSNA (Navarra Institute for Health Research), University of Navarra, E-31008 Pamplona, Navarra, Spain; (A.V.); (C.F.-R.); (J.P.-G.)
| | - Celia Fernández-Rubio
- Department of Microbiology and Parasitology, ISTUN Institute of Tropical Health, IdiSNA (Navarra Institute for Health Research), University of Navarra, E-31008 Pamplona, Navarra, Spain; (A.V.); (C.F.-R.); (J.P.-G.)
| | - Esther Larrea
- ISTUN Institute of Tropical Health, Navarra Institute for Health Research (IdiSNA), University of Navarra, E-31008 Pamplona, Navarra, Spain;
| | - José Peña-Guerrero
- Department of Microbiology and Parasitology, ISTUN Institute of Tropical Health, IdiSNA (Navarra Institute for Health Research), University of Navarra, E-31008 Pamplona, Navarra, Spain; (A.V.); (C.F.-R.); (J.P.-G.)
| | - Paul A. Nguewa
- Department of Microbiology and Parasitology, ISTUN Institute of Tropical Health, IdiSNA (Navarra Institute for Health Research), University of Navarra, E-31008 Pamplona, Navarra, Spain; (A.V.); (C.F.-R.); (J.P.-G.)
- Correspondence: ; Tel.: +34-948-425-600 (ext. 6434)
| |
Collapse
|
3
|
Ulusan Ö, Mert U, Sadıqova A, Öztürk S, Caner A. Identification of gene expression profiles in Leishmania major infection by integrated bioinformatics analyses. Acta Trop 2020; 208:105517. [PMID: 32360239 DOI: 10.1016/j.actatropica.2020.105517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 10/24/2022]
Abstract
Gene expression profiling in mouse models of leishmaniasis has given useful information to understand the molecular pathways active in lesions and to discover new diagnostic/therapeutic targets. Although the host response plays a critical role in protection from leishmaniasis and promoting disease severity, there are still unexplained aspects in the mechanism of non-healing cutaneous lesions, which need biomarkers for both targeted- therapy and diagnosis. To address this, transcriptional profiling of the skin lesions obtained from BALB/c mice infected with Leishmania major and healthy skin from naïve mice were evaluated by bioinformatics analysis, and then the results were validated by Revers Transcriptase-PCR. Five genes among the up-regulated differentially expressed genes named FCGR4, CCL4, CXCL9, Arg1 and IL-1β were found to have relatively high diagnostic value for CL due to L. major. Pathway analysis revealed that Triggering Receptor Expressed On Myeloid Cells 1 (TREM1) signaling pathways are active in cutaneous lesions, providing new insights for the understanding and treatment of leishmaniasis.
Collapse
Affiliation(s)
- Özlem Ulusan
- Department of Parasitology, Ege University Medical School, Izmir, Turkey
| | - Ufuk Mert
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Aygül Sadıqova
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Sercan Öztürk
- Departments of Computer Science, Dokuz Eylül University, Izmir, Turkey
| | - Ayse Caner
- Department of Parasitology, Ege University Medical School, Izmir, Turkey; Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey; Department of Bioinformatics, Ege University, Institute of Health Sciences, Izmir, Turkey; Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Heterogeneity of humoral immune response to Leishmania tropica in an experimental model. Parasitol Res 2019; 118:1231-1237. [PMID: 30778754 DOI: 10.1007/s00436-019-06256-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 02/11/2019] [Indexed: 10/27/2022]
Abstract
Humoral (antibody) response is an important part of immunity against pathogens. Despite the clear role of cell-mediated immune response in protection against leishmaniasis, the role of humoral responses is challenging. There is very limited data regarding humoral immune response against Leishmania tropica which is the causative agent of human cutaneous leishmaniasis in many parts of the world. Here, we have compared pathogenicity and antibody response against six Iranian Leishmania tropica isolates in BALB/c mice. A Leishmania major isolate was used for comparison. The parasites were injected into the mice followed by the evaluation of the lesion development, parasite load, and antibody responses (IgG1 and IgG2a). Our findings showed that some isolates caused the large lesions and high parasite load in the spleen and lymph node, while other isolates led to no lesion, no splenic parasitism, and low parasite load in the lymph node. The more pathogenic isolates induced higher antibody responses (IgG1 and IgG2a). Our results indicated that there is substantial heterogeneity among various Leishmania tropica isolates regarding the humoral immune response as well as the pathogenicity.
Collapse
|
5
|
Wieland A, Ahmed R. Fc Receptors in Antimicrobial Protection. Curr Top Microbiol Immunol 2019; 423:119-150. [DOI: 10.1007/82_2019_154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
6
|
Rostamian M, Niknam HM. Leishmania tropica: What we know from its experimental models. ADVANCES IN PARASITOLOGY 2018; 104:1-38. [PMID: 31030767 DOI: 10.1016/bs.apar.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Leishmania tropica causes different forms of leishmaniasis in many parts of the world. Animal models can help to clarify the issues of pathology and immune response in L. tropica infections and can be applied to the control, prevention and treatment of the disease. The aim of this article is to summarize published data related to experimental models of this parasite, presenting an overview of the subject. We also present in brief the epidemiology, transmission and human manifestation of L. tropica infection. Mice, rats and hamsters have been used for experimental models of L. tropica infection. Main findings of the published studies show that: (1) Hamsters are the best animal model for L. tropica infection, with the drawback of being outbred hence not suitable for many studies. (2) L. tropica infection causes a non-ulcerative and chronic pathology as cutaneous form in mice and usually visceral form in hamsters. (3) L. tropica infection in mice results in a weaker immune response in comparison to Leishmania major. (4) While the Th1 responses are evoked against L. tropica, Th2 responses do not explain the outcomes of this infection, and IL-10 and TGF-β are two main suppressive cytokines. (5) The host genotype affects the immune response and disease outcome of L. tropica infection and the dose, strain, routes of inoculation, and sex of the host are among the factors affecting disease outcome of this species.
Collapse
Affiliation(s)
- Mosayeb Rostamian
- Nosocomial Infections Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamid M Niknam
- Immunology Department, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
7
|
Martínez-López M, Soto M, Iborra S, Sancho D. Leishmania Hijacks Myeloid Cells for Immune Escape. Front Microbiol 2018; 9:883. [PMID: 29867798 PMCID: PMC5949370 DOI: 10.3389/fmicb.2018.00883] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/17/2018] [Indexed: 12/23/2022] Open
Abstract
Protozoan parasites of the Leishmania genus are the causative agents of leishmaniasis, a group of neglected tropical diseases whose clinical manifestations vary depending on the infectious Leishmania species but also on host factors. Recognition of the parasite by host myeloid immune cells is a key to trigger an effective Leishmania-specific immunity. However, the parasite is able to persist in host myeloid cells by evading, delaying and manipulating host immunity in order to escape host resistance and ensure its transmission. Neutrophils are first in infiltrating infection sites and could act either favoring or protecting against infection, depending on factors such as the genetic background of the host or the parasite species. Macrophages are the main host cells where the parasites grow and divide. However, macrophages are also the main effector population involved in parasite clearance. Parasite elimination by macrophages requires the priming and development of an effector Th1 adaptive immunity driven by specific subtypes of dendritic cells. Herein, we will provide a comprehensive outline of how myeloid cells regulate innate and adaptive immunity against Leishmania, and the mechanisms used by the parasites to promote their evasion and sabotage. Understanding the interactions between Leishmania and the host myeloid cells may lead to the development of new therapeutic approaches and improved vaccination to leishmaniases, an important worldwide health problem in which current therapeutic or preventive approaches are limited.
Collapse
Affiliation(s)
- María Martínez-López
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain
| | - Manuel Soto
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Salvador Iborra
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain.,Department of Immunology, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain
| |
Collapse
|
8
|
Abstract
Dengue provides the most abundant example in human medicine and the greatest human illness burden caused by the phenomenon of intrinsic antibody-dependent infection enhancement (iADE). In this immunopathological phenomenon infection of monocytes or macrophages using infectious immune complexes suppresses innate antiviral systems, permitting logarithmic intracellular growth of dengue virus. The four dengue viruses evolved from a common ancestor yet retain similar ecology and pathogenicity, but although infection with one virus provides short-term cross-protection against infection with a different type, millions of secondary dengue infections occur worldwide each year. When individuals are infected in the virtual absence of cross-protective dengue antibodies, the dengue vascular permeability syndrome (DVPS) may ensue. This occurs in around 2 to 4% of second heterotypic dengue infections. A complete understanding of the biologic mechanism of iADE, dengue biology, and the mechanism of host responses to dengue infection should lead to a comprehensive and complete understanding of the pathogenesis of DVPS. A crucial emphasis must be placed on understanding ADE. Clinical and epidemiological observations of DVPS define the research questions and provide research parameters. This article will review knowledge related to dengue ADE and point to areas where there has been little research progress. These observations relate to the two stages of dengue illnesses: afferent phenomena are those that promote the success of the microorganism to infect and survive; efferent phenomena are those mounted by the host to inhibit infection and replication and to eliminate the infectious agent and infected tissues. Data will be discussed as "knowns" and "unknowns."
Collapse
|
9
|
Fc Receptors and Fc Receptor-Like Molecules within the Immunoreceptor Family. ENCYCLOPEDIA OF IMMUNOBIOLOGY 2016. [PMCID: PMC7152311 DOI: 10.1016/b978-0-12-374279-7.02017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Receptors for the Fc portion of immunoglobulins (FcRs) account for most cell-mediated biological activities of antibodies. The majority of FcRs are encoded by a set of genes, clustered in the fcr locus, on chromosome 1 in humans and on chromosome 1 and 3 in mice. Eight (in humans) and six (in mice) new genes were found, intermixed with FcR genes in corresponding fcr loci, which encode FcR-like molecules (FcRLs). FcRs and FcRLs are genetically, phylogenetically, structurally, and functionally related. FcRs and FcRLs, however, markedly differ by their ligands, their tissue distribution, and, therefore, by the biological functions they control. A systematic comparison of their biological properties leads to the conclusion that FcRLs are not like FcRs. They altogether form a single family within the immunoreceptor family, whose members fulfill distinct but complementary roles in immunity by differentially controlling innate and adaptive responses.
Collapse
|
10
|
Synergistic Use of Geniposide and Ginsenoside Rg1 Balance Microglial TNF-α and TGF-β1 following Oxygen-Glucose Deprivation In Vitro: A Genome-Wide Survey. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:756346. [PMID: 26693244 PMCID: PMC4677035 DOI: 10.1155/2015/756346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 10/02/2015] [Accepted: 10/15/2015] [Indexed: 12/12/2022]
Abstract
Ischemia-activated microglia are like a double-edged sword, characterized by both neurotoxic and neuroprotective effects. The aim of this study was to reveal the synergistic effect of geniposide and ginsenoside Rg1 based on tumor necrosis factor- (TNF-) α and transforming growth factor- (TGF-) β1 balance of microglia. BV2 microglial cells were divided into 5 groups: control, model (oxygen-glucose deprivation (OGD)), geniposide-treated, ginsenoside-Rg1-treated, and combination-treated. A series of assays were used to detect on (i) cell viability; (ii) NO content; (iii) expression (content) of TNF-α and TGF-β1; and (iv) gene expression profiles. The results showed that integrated use of geniposide and ginsenoside Rg1 significantly inhibited NO level and protected cell viability, improved the content and expression of TGF-β1, and reduced the content and expression of TNF-α. Separated use of geniposide or ginsenoside Rg1 showed different effects at different emphases. Next-generation sequencing showed that Fcγ-receptor-mediated phagocytosis pathway played a key regulatory role in the balance of TNF-α and TGF-β1 when cotreated with geniposide and ginsenoside Rg1. These findings suggest that synergistic drug combination of geniposide and ginsenoside Rg1 in the treatment of stroke is a feasible avenue for the application.
Collapse
|
11
|
Rostamian M, Sohrabi S, Kavosifard H, Niknam HM. Lower levels of IgG1 in comparison with IgG2a are associated with protective immunity against Leishmania tropica infection in BALB/c mice. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2015; 50:160-166. [PMID: 26066544 DOI: 10.1016/j.jmii.2015.05.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 04/06/2015] [Accepted: 05/05/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND/PURPOSE Leishmania (L.) tropica is the causative agent of different forms of human leishmaniasis. There is little information about the role of Leishmania-specific antibodies in the immune response against L. tropica infection. The aim of this study is to evaluate the role of Leishmania-specific antibodies and their immunoglobulin G (IgG) isotypes in L. tropica infection. METHODS L. tropica at two different doses (high dose, 106 parasites/mouse and low dose, 103 parasites/mouse) were used for infection of BALB/c mice. BALB/c mice infected with Leishmania major were used for comparison. Anti-Leishmania antibodies of the IgG1 and IgG2a isotypes were assayed by enzyme-linked immunosorbent assay. RESULTS Our data showed that (1) a higher parasite dose results in higher levels of antibody. (2) L. tropica infection results in a lower IgG1 antibody response, compared with L. major infection. (3) The IgG2a/IgG1 antibody response in L. tropica infection is higher than that in L. major infection. CONCLUSION A higher IgG2a/IgG1 ratio is associated with protective immune response in L. tropica infection. These data can help to approach the complex profile of immunity against L. tropica infection.
Collapse
Affiliation(s)
| | - Saeed Sohrabi
- Department of Microbiology, Qom Branch of Islamic Azad University, Qom, Iran
| | - Hanie Kavosifard
- Department of Microbiology, Kurdistan Science and Research Branch, Islamic Azad University, Sanandaj, Iran
| | - Hamid M Niknam
- Immunology Department, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
12
|
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
|
13
|
de Matos Guedes HL, da Silva Costa BL, Chaves SP, de Oliveira Gomes DC, Nosanchuk JD, De Simone SG, Rossi-Bergmann B. Intranasal vaccination with extracellular serine proteases of Leishmania amazonensis confers protective immunity to BALB/c mice against infection. Parasit Vectors 2014; 7:448. [PMID: 25239157 PMCID: PMC4261548 DOI: 10.1186/1756-3305-7-448] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 09/01/2014] [Indexed: 12/12/2022] Open
Abstract
Background Previously, we demonstrated that unlike subcutaneous or intramuscular vaccination, intranasal vaccination of BALB/c mice with whole Leishmania amazonensis antigens leads to protection against cutaneous leishmaniasis. Here, the role of parasite serine proteases in the protective immunity was investigated. Findings Serine Proteases were partially purified from both soluble (LaSP-Sol) and extracellular (LaSP-Ex) Leishmania amazonensis promastigote extracts by aprotinin-agarose chromatography. BALB/c mice were intranasally immunized with LaSP-Sol and LaSP-Ex prior to infection with L. amazonensis. LaSP-Ex but not LaSP-Sol vaccination led to significantly smaller lesions and parasite burdens as compared with non-vaccinated controls. Protection was accompanied by systemic Th1 polarization with increased IFN-γ and decreased IL-4 and IL-10 splenic production. Likewise, increased production of IFN-γ, IL-12 and IL-4 concomitant with decreased TGF-β and TNF-α was locally observed in the infected footpad. Conclusion This study indicates that extracellular serine proteases of L. amazonensis are strong candidates for a more defined intranasal vaccine against cutaneous leishmaniasis. Electronic supplementary material The online version of this article (doi:10.1186/1756-3305-7-448) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Herbert Leonel de Matos Guedes
- Laboratório de Imunofarmacologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | | | |
Collapse
|
14
|
Vitry MA, Hanot Mambres D, De Trez C, Akira S, Ryffel B, Letesson JJ, Muraille E. Humoral Immunity and CD4+Th1 Cells Are Both Necessary for a Fully Protective Immune Response upon Secondary Infection withBrucella melitensis. THE JOURNAL OF IMMUNOLOGY 2014; 192:3740-52. [DOI: 10.4049/jimmunol.1302561] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
15
|
Sousa LMA, Carneiro MBH, Resende ME, Martins LS, Dos Santos LM, Vaz LG, Mello PS, Mosser DM, Oliveira MAP, Vieira LQ. Neutrophils have a protective role during early stages of Leishmania amazonensis infection in BALB/c mice. Parasite Immunol 2014; 36:13-31. [PMID: 24102495 DOI: 10.1111/pim.12078] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 08/05/2013] [Indexed: 01/13/2023]
Abstract
Neutrophils are involved in the early stages of immune responses to pathogens. Here, we investigated the role of neutrophils during the establishment of Leishmania amazonensis infection in BALB/c and C57BL/6 mice. First, we showed an accumulation of neutrophils between 6 and 24 h post-infection, followed by a reduction in neutrophil numbers after 72 h. Next, we depleted neutrophils prior to infection using RB6-8C5 or 1A8 mAb. Neutrophil depletion led to faster lesion development, increased parasite numbers and higher arginase activity during the first week of infection in BALB/c mice, but not in C57BL/6 mice. Increased susceptibility was accompanied by augmented levels of anti-L. amazonensis IgG and increased production of IL-10 and IL-17. Because IL-10 is a mediator of susceptibility to Leishmania infection, we blocked IL-10 signalling in neutrophil-depleted mice using anti-IL-10R. Interestingly, inhibition of IL-10 signalling abrogated the increase in parasite loads observed in neutrophil-depleted mice, suggesting that parasite proliferation is at least partially mediated by IL-10. Additionally, we tested the effect of IL-17 in inflammatory macrophages and observed that IL-17 increased arginase activity and favoured parasite growth. Taken together, our data indicate that neutrophils control parasite numbers and limit lesion development during the first week of infection in BALB/c mice.
Collapse
Affiliation(s)
- L M A Sousa
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Guilliams M, Bruhns P, Saeys Y, Hammad H, Lambrecht BN. The function of Fcγ receptors in dendritic cells and macrophages. Nat Rev Immunol 2014; 14:94-108. [PMID: 24445665 DOI: 10.1038/nri3582] [Citation(s) in RCA: 484] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) and macrophages use various receptors to recognize foreign antigens and to receive feedback control from adaptive immune cells. Although it was long believed that all immunoglobulin Fc receptors are universally expressed by phagocytes, recent findings indicate that only monocyte-derived DCs and macrophages express high levels of activating Fc receptors for IgG (FcγRs), whereas conventional and plasmacytoid DCs express the inhibitory FcγR. In this Review, we discuss how the uptake, processing and presentation of antigens by DCs and macrophages is influenced by FcγR recognition of immunoglobulins and immune complexes in the steady state and during inflammation.
Collapse
Affiliation(s)
- Martin Guilliams
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Pierre Bruhns
- 1] Institut Pasteur, Département d'Immunologie, Laboratoire Anticorps en Thérapie et Pathologie, 75015 Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U760, 75015 Paris, France
| | - Yvan Saeys
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Hamida Hammad
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium. [3] Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| |
Collapse
|
17
|
Abstract
Antibodies are major molecular effectors of adaptive immune responses. Most, if not all, biological activities of antibodies, however, depend on the functional properties of cells that express receptors for the Fc portion of antibodies (FcR). Most FcR are activating receptors; some are inhibitory. When engaged by antibodies and antigen, the various FcR expressed by a given cell trigger a mixture of positive and negative signals whose integration determines cellular responses. Responses of cell populations can be either protective or pathogenic. As a consequence, FcR are potential target/tools in a variety of diseases including infection, allergy, autoimmune diseases, and cancer.
Collapse
|
18
|
Abstract
Most biological activities of antibodies depend on their ability to engage Receptors for the Fc portion of immunoglobulins (FcRs) on a variety of cell types. As FcRs can trigger positive and negative signals, as these signals control several biological activities in individual cells, as FcRs are expressed by many cells of hematopoietic origin, mostly of the myeloid lineage, as these cells express various combinations of FcRs, and as FcR-expressing cells have different functional repertoires, antibodies can exert a wide spectrum of biological activities. Like B and T Cell Receptors (BCRs and TCRs), FcRs are bona fide immunoreceptors. Unlike BCRs and TCRs, however, FcRs are immunoreceptors with an adaptive specificity for antigen, with an adaptive affinity for antibodies, with an adaptive structure and with an adaptive signaling. They induce adaptive biological responses that depend on their tissue distribution and on FcR-expressing cells that are selected locally by antibodies. They critically determine health and disease. They are thus exquisitely adaptive therapeutic tools.
Collapse
Affiliation(s)
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
19
|
Absence of FcγRIII results in increased proinflammatory response in FcγRIII-KO cardiac recipients. Transplantation 2013; 96:601-8. [PMID: 23903009 DOI: 10.1097/tp.0b013e31829c2455] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Alloantibody can contribute significantly to rejection of heart transplants by activation of complement and interactions with a variety of effector cells, including macrophages and monocytes through activating FcγRI, FcγRIII, FcγRIV, the inhibitory FcγRIIB and complement receptors. These receptors link cellular and humoral immunity by bridging the antibody specificity to effector cells. Activating FcγRs are also involved in serum amyloid P component (SAP)-mediated clearance of apoptotic bodies. METHODS B10.A (H-2a) hearts were transplanted into wild-type (WT) or FcγRIII-knockout (KO) C57BL/6 (H-2b) mouse recipients. Levels of alloantibodies and SAP in the circulation were determined by flow cytometry and enzyme-linked immunosorbent assay, respectively. Intragraft cytokine mRNA expression was measured by real-time polymerase chain reaction. Intragraft deposition of C4d, von Willebrand factor, SAP, and activated caspase 3 was visualized by immunochemistry. RESULTS B10.A hearts in C57BL/6 FcγRIII-KO recipients were rejected acutely within 6 to 8 days compared with 10 to 14 days in WT. The rejection in FcγRIII-KO was accompanied by higher levels of circulating IgM/IgG alloantibodies and SAP than in WT recipients. Histology in FcγRIII-KO cardiac allograft recipients indicated perivascular margination of monocytes and neutrophils, vascular endothelial cell injury, and intense vasculocentric infiltrates with extensive apoptosis. Higher numbers of apoptotic cells, stronger C4d and SAP deposition, and extensive activated caspase 3 were found in areas of dense pockets of apoptotic blebs in FcγRIII-KO. CONCLUSIONS We propose that absence of FcγRIII is associated with the lack of efficient SAP-mediated clearance of apoptotic cells through FcγRs. Apoptotic cells become immunogenic and induce enhanced inflammation, alloantibody production, and complement activation leading to accelerated cardiac allograft rejection.
Collapse
|
20
|
Srivastava S, Pandey SP, Jha MK, Chandel HS, Saha B. Leishmania expressed lipophosphoglycan interacts with Toll-like receptor (TLR)-2 to decrease TLR-9 expression and reduce anti-leishmanial responses. Clin Exp Immunol 2013; 172:403-9. [PMID: 23600828 DOI: 10.1111/cei.12074] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2013] [Indexed: 12/12/2022] Open
Abstract
Two different Toll-like receptors (TLRs) have been shown to play a role in host responses to Leishmania infection. TLR-2 is involved in parasite survival in macrophages upon activation by lipophosphoglycan (LPG), a virulence factor expressed by Leishmania. In contrast, activation of TLR-9 has been shown to promote a host-protective response. However, whether there is a relationship between the interaction of LPG and TLR-2, on one hand, with the effect of TLR-9, on the other hand, remains unknown. In this study, we report that in-vitro infection of macrophages with a L. major parasite with high expression levels of LPG results in decreased TLR-9 expression compared to infection with a L. major parasite with lower expression levels of LPG. Addition of anti-LPG as well as anti-TLR-2 antibodies prevents this reduction of TLR-9 expression. Also, the addition of purified LPG to macrophages results in a decrease of TLR-9 expression, which is shown to be mediated by transforming growth factor (TGF)-β and interleukin (IL)-10. Finally, in-vitro treatment of macrophages with anti-LPG and/or anti-TLR-2 antibodies before infection reduces the number of amastigotes in macrophages and co-treatment of mice with anti-TLR-2 antibodies and cytosine-phosphate-guanosine (CpG) reduces footpad swelling and parasite load in the draining lymph nodes, accompanied by an interferon (IFN)-γ-predominant T cell response. Thus, for the first time, we show how interactions between LPG and TLR-2 reduce anti-leishmanial responses via cytokine-mediated decrease of TLR-9 expression.
Collapse
|
21
|
Resende M, Moreira D, Augusto J, Cunha J, Neves B, Cruz MT, Estaquier J, Cordeiro-da-Silva A, Silvestre R. Leishmania-infected MHC class IIhigh dendritic cells polarize CD4+ T cells toward a nonprotective T-bet+ IFN-γ+ IL-10+ phenotype. THE JOURNAL OF IMMUNOLOGY 2013; 191:262-73. [PMID: 23729437 DOI: 10.4049/jimmunol.1203518] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A differential behavior among infected and bystander dendritic cells (DCs) has been explored in different infection models. We have analyzed both populations sorted on contact with visceral Leishmania infantum on a susceptible mice model evaluating the subsequent repercussions on adaptive immune response. Our results demonstrate a clear dichotomy between the immunomodulatory abilities of bystander and infected DCs. The bystander population presents increased levels of IL-12p40 and costimulatory molecules being capable to induce CD4(+) T cell activation with immune protective capabilities. In contrast, infected DCs, which express lower costimulatory molecules and higher levels of IL-10, promote the development of Leishmania Ag-specific, nonprotective T-bet(+)IFN-γ(+)IL-10(+) CD4(+) T cells with an effector phenotype. This specific polarization was found to be dependent on IL-12p70. Splenic infected DCs recovered from chronic infected animals are similarly capable to polarize ex vivo syngeneic naive CD4(+) T cells toward a T-bet(+)IFN-γ(+)IL-10(+) phenotype. Further analysis revealed that only MHC class II(high)-infected DCs were responsible for this polarization. The adoptive transfer of such polarized CD4(+) T cells facilitates visceral leishmaniasis in BALB/c mice in a clear contrast with their counterpart generated with bystander DCs that significantly potentiate protection. Further, we demonstrated that CD4(+) T cells primed by infected DCs in an IL-10 free system, thus deprived of T-bet(+)IFN-γ(+)IL-10(+) population, restore the immune response and reduce parasite load, supporting a deleterious role of IFN-γ(+)IL-10(+) T cells in the maintenance of infection. Overall, our results highlight novel subversion mechanisms by which nonprotective T-bet(+)IFN-γ(+)IL-10(+) T cells are associated with chronicity and prolonged parasite persistence.
Collapse
Affiliation(s)
- Mariana Resende
- Parasite Disease Group, Institute for Molecular and Cell Biology, University of Porto, 4150-180 Porto, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Polando R, Dixit UG, Carter CR, Jones B, Whitcomb JP, Ballhorn W, Harintho M, Jerde CL, Wilson ME, McDowell MA. The roles of complement receptor 3 and Fcγ receptors during Leishmania phagosome maturation. J Leukoc Biol 2013; 93:921-32. [PMID: 23543768 DOI: 10.1189/jlb.0212086] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Leishmania are intracellular parasites adapted to surviving in macrophages, whose primary function is elimination of invading pathogens. Leishmania entry into host cells is receptor-mediated. These parasites are able to engage multiple host cell-surface receptors, including MR, TLRs, CR3, and FcγRs. Here, we investigated the role of CR3 and FcγR engagement on the maturation of Leishmania-containing phagosomes using CD11b-/- and FcγR-/- macrophages, and assessing EEA1 and lysosome-associated proteins is necessary for the phagosome maturation delay, characteristic of Leishmania infection. Leishmania-containing phagosomes do not fuse with lyosomes until 5 h postinfection in WT mice. Phagolysosome fusion occurs by 1 h in CD11b and FcγR common chain KO macrophages, although receptor deficiency does not influence Leishmania entry or viability. We also investigated the influence of serum components and their effects on phagosome maturation progression. Opsonization with normal mouse serum, complement-deficient serum, or serum from Leishmania-infected mice all influenced phagosome maturation progression. Our results indicate that opsonophagocytosis influences phagosomal trafficking of Leishmania without altering the intracellular fate.
Collapse
Affiliation(s)
- Rachel Polando
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Alexander J, Brombacher F. T helper1/t helper2 cells and resistance/susceptibility to leishmania infection: is this paradigm still relevant? Front Immunol 2012; 3:80. [PMID: 22566961 PMCID: PMC3342373 DOI: 10.3389/fimmu.2012.00080] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 03/28/2012] [Indexed: 11/13/2022] Open
Abstract
Work in large part on Leishmania major in the 1980s identified two distinct apparently counter-regulatory CD4+ T cell populations, T helper (h)1 and Th2, that controlled resistance/susceptibility to infection respectively. However, the generation of IL-4−/− mice in the 1990s questioned the paramount role of this Th2 archetypal cytokine in the non-healing response to Leishmania infection. The more recent characterization of CD4+ T cell regulatory populations and further effector CD4+ T helper populations, Th17, Th9, and T follicular (f)h cells as well as the acknowledged plasticity in T helper cell function has further added to the complexity of host pathogen interactions. These interactions are complicated by the multiplicity of cells that respond to CD4+ T cell subset signatory cytokines, as well as the diversity of Leishmania species that are often subject to significantly different immune-regulatory controls. In this article we review current knowledge with regard to the role of CD4+ T cells and their products during Leishmania infection. In particular we update on our studies using conditional IL-4Rα gene-deficient mice that have allowed dissection of the cell interplay dictating the disease outcomes of the major Leishmania species infecting humans.
Collapse
Affiliation(s)
- James Alexander
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde Glasgow, UK
| | | |
Collapse
|
24
|
Iyer SS, Cheng G. Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit Rev Immunol 2012; 32:23-63. [PMID: 22428854 DOI: 10.1615/critrevimmunol.v32.i1.30] [Citation(s) in RCA: 983] [Impact Index Per Article: 75.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Interleukin 10 (IL-10) is a cytokine with potent anti-inflammatory properties that plays a central role in limiting host immune response to pathogens, thereby preventing damage to the host and maintaining normal tissue homeostasis. Dysregulation of IL-10 is associated with enhanced immunopathology in response to infection as well as increased risk for development of many autoimmune diseases. Thus a fundamental understanding of IL-10 gene expression is critical for our comprehension of disease progression and resolution of host inflammatory response. In this review, we discuss modes of regulation of IL-10 gene expression in immune effector cell types, including signal transduction, epigenetics, promoter architecture, and post-transcriptional regulation, and how aberrant regulation contributes to immunopathology and disease progression.
Collapse
|
25
|
de Assis RR, Ibraim IC, Nogueira PM, Soares RP, Turco SJ. Glycoconjugates in New World species of Leishmania: polymorphisms in lipophosphoglycan and glycoinositolphospholipids and interaction with hosts. Biochim Biophys Acta Gen Subj 2011; 1820:1354-65. [PMID: 22093608 DOI: 10.1016/j.bbagen.2011.11.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 10/31/2011] [Accepted: 11/01/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND Protozoan parasites of the genus Leishmania cause a number of important diseases in humans and undergo a complex life cycle, alternating between a sand fly vector and vertebrate hosts. The parasites have a remarkable capacity to avoid destruction in which surface molecules are determinant for survival. Amongst the many surface molecules of Leishmania, the glycoconjugates are known to play a central role in host-parasite interactions and are the focus of this review. SCOPE OF THE REVIEW The most abundant and best studied glycoconjugates are the Lipophosphoglycans (LPGs) and glycoinositolphospholipids (GIPLs). This review summarizes the main studies on structure and biological functions of these molecules in New World Leishmania species. MAJOR CONCLUSIONS LPG and GIPLs are complex molecules that display inter- and intraspecies polymorphisms. They are key elements for survival inside the vector and to modulate the vertebrate immune response during infection. GENERAL SIGNIFICANCE Most of the studies on glycoconjugates focused on Old World Leishmania species. Here, it is reported some of the studies involving New World species and their biological significance on host-parasite interaction. This article is part of a Special Issue entitled Glycoproteomics.
Collapse
Affiliation(s)
- Rafael Ramiro de Assis
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, FIOCRUZ, Av. Augusto de Lima, 1715, Belo Horizonte, MG 30190-002, Brazil
| | | | | | | | | |
Collapse
|
26
|
Oliveira CR, Pereira LIA, Pereira AJCS, Ferreira AA, Crespo AMC, Silveira LA. Allelic polymorphism of human FcγRIIA-H/R131 receptor in American tegumentary leishmaniasis. Int J Immunogenet 2011; 38:225-31. [DOI: 10.1111/j.1744-313x.2011.00997.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
Okwor I, Uzonna JE. Immunotherapy as a strategy for treatment of leishmaniasis: a review of the literature. Immunotherapy 2011; 1:765-76. [PMID: 20636022 DOI: 10.2217/imt.09.40] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Leishmaniasis occurs as a spectrum of clinical syndromes divided into cutaneous, mucocutaneous and visceral forms. The epidemiology and clinical features are highly variable owing to the interplay of many factors ranging from parasite species and strains, vectors, host genetics and environment. Currently, there is no effective licensed vaccine for use in humans against leishmaniasis. Most traditional and low-cost treatment options, particularly in poor and endemic areas, are toxic with many adverse reactions and they require a long course of administration. The use of more effective, less toxic drugs is limited because total treatment cost is very high (expensive) and there are fears of development of drug resistance. Recent studies indicate that certain strategies aimed at modulating the host immune response (collectively called immunotherapy) could result in prophylactic and/or therapeutic cure of leishmaniasis under both laboratory and field conditions. In this review, we focus on treatment of leishmaniasis with a particular emphasis on immunotherapy/immunochemotherapy as an alternative to conventional drug treatment.
Collapse
Affiliation(s)
- Ifeoma Okwor
- Parasite Vaccines Development Laboratory, Department of Immunology, Faculty of Medicine, University of Manitoba, 750 McDermot Avenue, Winnipeg, Manitoba R3E 0W3, Canada
| | | |
Collapse
|
28
|
Stäger S, Joshi T, Bankoti R. Immune evasive mechanisms contributing to persistent Leishmania donovani infection. Immunol Res 2010; 47:14-24. [PMID: 20087685 DOI: 10.1007/s12026-009-8135-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The protozoan parasite Leishmania donovani, a causative agent of visceral leishmaniasis, has evolved several strategies to interfere with the immune system and establish persistent infections that are potentially lethal. In this article, we discuss two mechanisms of immune evasion adopted by the parasite: the induction of immune suppressive IL-10 responses and the generation of poor and functionally impaired CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Simona Stäger
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, BRB, Rm 655, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
29
|
Hedrich CM, Bream JH. Cell type-specific regulation of IL-10 expression in inflammation and disease. Immunol Res 2010; 47:185-206. [PMID: 20087682 DOI: 10.1007/s12026-009-8150-5] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
IL-10 plays an essential part in controlling inflammation and instructing adaptive immune responses. Consequently, dysregulation of IL-10 is linked with susceptibility to numerous infectious and autoimmune diseases in mouse models and in humans. It has become increasingly clear that appropriate temporal/spatial expression of IL-10 may be the key to how IL-10 contributes to the delicate balance between inflammation and immunoregulation. The mechanisms that govern the cell type- and receptor-specific induction of IL-10, however, remain unclear. This is due largely to the wide distribution of cellular sources that express IL-10 under diverse stimulation conditions and in a variety of tissue compartments. Further complicating the issue is the fact that human IL-10 expression patterns appear to be under genetic influence resulting in differential expression and disease susceptibility. In this review, we discuss the cellular sources of IL-10, their link to disease phenotypes and the molecular mechanisms implicated in IL-10 regulation.
Collapse
Affiliation(s)
- Christian M Hedrich
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Room E5624, Baltimore, MD 21205-1901, USA
| | | |
Collapse
|
30
|
Halstead SB, Mahalingam S, Marovich MA, Ubol S, Mosser DM. Intrinsic antibody-dependent enhancement of microbial infection in macrophages: disease regulation by immune complexes. THE LANCET. INFECTIOUS DISEASES 2010; 10:712-22. [PMID: 20883967 PMCID: PMC3057165 DOI: 10.1016/s1473-3099(10)70166-3] [Citation(s) in RCA: 298] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A wide range of microorganisms can replicate in macrophages, and cell entry of these pathogens via non-neutralising IgG antibody complexes can result in increased intracellular infection through idiosyncratic Fcγ-receptor signalling. The activation of Fcγ receptors usually leads to phagocytosis. Paradoxically, the ligation of monocyte or macrophage Fcγ receptors by IgG immune complexes, rather than aiding host defences, can suppress innate immunity, increase production of interleukin 10, and bias T-helper-1 (Th1) responses to Th2 responses, leading to increased infectious output by infected cells. This intrinsic antibody-dependent enhancement (ADE) of infection modulates the severity of diseases as disparate as dengue haemorrhagic fever and leishmaniasis. Intrinsic ADE is distinct from extrinsic ADE, whereby complexes of infectious agents with non-neutralising antibodies lead to an increased number of infected cells. Intrinsic ADE might be involved in many protozoan, bacterial, and viral infections. We review insights into intracellular mechanisms and implications of enhanced pathogenesis after ligation of macrophage Fcγ receptors by infectious immune complexes.
Collapse
|
31
|
Goto Y, Carter D, Guderian J, Inoue N, Kawazu SI, Reed SG. Upregulated expression of B-cell antigen family tandem repeat proteins by Leishmania amastigotes. Infect Immun 2010; 78:2138-45. [PMID: 20160013 PMCID: PMC2863543 DOI: 10.1128/iai.01102-09] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 11/10/2009] [Accepted: 02/10/2010] [Indexed: 12/18/2022] Open
Abstract
Proteins with tandem repeat (TR) domains have been found in various protozoan parasites, and they are often targets of B-cell responses. Through systematic analyses of whole proteomes, we recently demonstrated that two trypanosomatid parasites, Leishmania infantum and Trypanosoma cruzi, are rich in antigenic proteins with large TR domains. However, the reason that these proteins are antigenic was unclear. Here, by performing molecular, immunological, and bioinformatic characterizations of Leishmania TR proteins, we found two possible factors affecting the antigenicity of these proteins; one factor is their fundamental composition as TR proteins, and the other is regulation of their expression by parasites. Enzyme-linked immunosorbent assays (ELISAs) using recombinant proteins revealed that the copy number of the repeat affects the affinity of binding between antigens and antibodies, as expected based on thermodynamic binding kinetics. Other than containing TR domains, the TR proteins do not share characteristics, such as sequence similarity or biased cellular location predicted by the presence of a signal sequence(s) and/or a transmembrane domain(s). However, the TR proteome contained a higher percentage of proteins upregulated in amastigotes than the whole proteome, and upregulated expression of a TR protein seemed to affect its antigenicity. These results indicate that Leishmania parasites actively utilize the TR protein family for parasitism in mammalian hosts.
Collapse
Affiliation(s)
- Yasuyuki Goto
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan.
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Much remains to be learned about the pathogenesis of the different manifestations of dengue virus (DENV) infections in humans. They may range from subclinical infection to dengue fever, dengue hemorrhagic fever (DHF), and eventually dengue shock syndrome (DSS). As both cell tropism and tissue tropism of DENV are considered major determinants in the pathogenesis of dengue, there is a critical need for adequate tropism assays, animal models, and human autopsy data. More than 50 years of research on dengue has resulted in a host of literature, which strongly suggests that the pathogenesis of DHF and DSS involves viral virulence factors and detrimental host responses, collectively resulting in abnormal hemostasis and increased vascular permeability. Differential targeting of specific vascular beds is likely to trigger the localized vascular hyperpermeability underlying DSS. A personalized approach to the study of pathogenesis will elucidate the basis of individual risk for development of DHF and DSS as well as identify the genetic and environmental bases for differences in risk for development of severe disease.
Collapse
|
33
|
Abstract
Genetic defects affecting the humoral immune response and especially the production of antibodies of the immunoglobulin G (IgG) isotype result in a heightened susceptibility to infections. Studies over the last years have demonstrated the crucial role of Fc-receptors for IgG (FcγRs) widely expressed on innate immune effector cells in mediating the protective function of IgG. During the last years, additional ligands interacting with FcγRs as well as additional receptors binding to IgG glycosylation variants have been identified. In this review, we discuss how the interaction of these different ligands with classical and novel Fcγ-receptors influences the immune response and which strategies microorganisms have developed to prevent them.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Chair of Genetics, University of Erlangen-Nuremberg, Staudtstr. 5, 91054, Erlangen, Germany.
| | | |
Collapse
|
34
|
Ronet C, Hauyon-La Torre Y, Revaz-Breton M, Mastelic B, Tacchini-Cottier F, Louis J, Launois P. Regulatory B cells shape the development of Th2 immune responses in BALB/c mice infected with Leishmania major through IL-10 production. THE JOURNAL OF IMMUNOLOGY 2009; 184:886-94. [PMID: 19966209 DOI: 10.4049/jimmunol.0901114] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent evidence indicates that B cells are required for susceptibility to infection with Leishmania major in BALB/c mice. In this study, we analyzed the role of the IL-10 produced by B cells in this process. We showed that B cells purified from the spleen of BALB/c mice produced IL-10 in response to stimulation with L. major in vitro. In vivo, early IL-10 mRNA expression is detected after L. major infection in B cells from draining lymph nodes of susceptible BALB/c, but not of resistant C57BL/6 mice. Although adoptive transfer of naive wild-type B cells prior to infection in B cell-deficient BALB/c mice restored Th2 cell development and susceptibility to infection with L. major of these otherwise resistant mice, adoptive transfer of IL-10(-/-) B cells mice did not. B cells stimulated by L. major, following in vitro or in vivo encounter, express the CD1d and CD5 molecules and the IL-10 produced by these cells downregulate IL-12 production by L. major-stimulated dendritic cells. These observations indicate that IL-10 secreting B cells are phenotypically and functionally regulatory B cells. Altogether these results demonstrate that the IL-10 produced by regulatory CD1d+ CD5+ B cells in response to L. major is critical for Th2 cell development in BALB/c mice.
Collapse
Affiliation(s)
- Catherine Ronet
- World Health Organization Immunology Research and Training Center, University of Lausanne, Epalinges, Switzerland
| | | | | | | | | | | | | |
Collapse
|
35
|
Kurey I, Kobets T, Havelková H, Slapnicková M, Quan L, Trtková K, Grekov I, Svobodová M, Stassen AP, Hutson A, Demant P, Lipoldová M. Distinct genetic control of parasite elimination, dissemination, and disease after Leishmania major infection. Immunogenetics 2009; 61:619-33. [PMID: 19705113 PMCID: PMC2744819 DOI: 10.1007/s00251-009-0392-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 07/27/2009] [Indexed: 10/28/2022]
Abstract
Elimination of pathogens is the basis of host resistance to infections; however, relationship between persisting pathogens and disease has not been clarified. Leishmania major infection in mice is an important model of host-pathogen relationship. Infected BALB/c mice exhibit high parasite numbers in lymph nodes and spleens, and a chronic disease with skin lesions, splenomegaly, and hepatomegaly, increased serum IgE levels and cytokine imbalance. Although numerous gene loci affecting these disease symptoms have been reported, genes controlling parasites' elimination or dissemination have never been mapped. We therefore compared genetics of the clinical and immunologic symptomatology with parasite load in (BALB/c x CcS-11) F2 hybrids and mapped five loci, two of which control parasite elimination or dissemination. Lmr5 influences parasite loads in spleens (and skin lesions, splenomegaly, and serum IgE, IL-4, and IFNgamma levels), and Lmr20 determines parasite numbers in draining lymph nodes (and serum levels of IgE and IFNgamma), but no skin or visceral pathology. Three additional loci do not affect parasite numbers but influence significantly the disease phenotype-Lmr21: skin lesions and IFNgamma levels, Lmr22: IL-4 levels, Lmr23: IFNgamma levels, indicating that development of L. major-caused disease includes critical regulations additional to control of parasite spread.
Collapse
Affiliation(s)
- Iryna Kurey
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídenská 1083, 142 20 Prague 4, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kamala T, Nanda NK. Protective response to Leishmania major in BALB/c mice requires antigen processing in the absence of DM. THE JOURNAL OF IMMUNOLOGY 2009; 182:4882-90. [PMID: 19342667 DOI: 10.4049/jimmunol.0803956] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Protection from the parasite Leishmania major is mediated by CD4 T cells. BALB/c mice are susceptible to L. major and show a nonprotective immunodominant CD4 T cell response to Leishmania homolog of activated receptor for c-kinase (LACK) 158-173. Host genes that underlie BALB/c susceptibility to L. major infections are poorly defined. DM, a nonclassical MHC class II molecule, due to its peptide editing properties has been shown to 1) edit the repertoire of peptides displayed by APC, and 2) focus the display of epitopes by APC to the immunodominant ones. We tested the hypothesis that deficiency of DM, by causing presentation of a different array of epitopes by infected APC than that presented by DM-sufficient APC, may change the course of L. major infection in the susceptible BALB/c mice. We show herein that unlike their susceptible wild-type counterparts, BALB/c mice deficient in DM are protected from infections with L. major. Furthermore, DM-deficient mice fail to display the immunodominant LACK 158-173 on infected APC. In its place, infected DM(-/-) hosts show elicitation of CD4 T cells specific for newer epitopes not presented by wild-type L. major-infected APC. Protection of BALB/c DM(-/-) mice is dependent on IFN-gamma. DM is thus a host susceptibility gene in BALB/c mice, and Ag processing in the absence of DM results in elicitation of a protective T cell response against L. major infections. This report suggests a novel mechanism to trigger host resistance against pathogens.
Collapse
Affiliation(s)
- Tirumalai Kamala
- Laboratory of Cellular and Molecular Immunology, T-Cell Tolerance and Memory Section, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
37
|
Ponte-Sucre A, Gulder T, Wegehaupt A, Albert C, Rikanović C, Schaeflein L, Frank A, Schultheis M, Unger M, Holzgrabe U, Bringmann G, Moll H. Structure−Activity Relationship and Studies on the Molecular Mechanism of Leishmanicidal N,C-Coupled Arylisoquinolinium Salts. J Med Chem 2008; 52:626-36. [DOI: 10.1021/jm801084u] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Alicia Ponte-Sucre
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Tanja Gulder
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Annemarie Wegehaupt
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Christoph Albert
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Carina Rikanović
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Leonhard Schaeflein
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Andreas Frank
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Martina Schultheis
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Matthias Unger
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Ulrike Holzgrabe
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Gerhard Bringmann
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| | - Heidrun Moll
- Institute of Molecular Infection Biology, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany, Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany, Laboratory of Molecular Physiology, Universidad Central de Venezuela, Caracas, Venezuela
| |
Collapse
|
38
|
Fc receptor gamma-chain, a constitutive component of the IL-3 receptor, is required for IL-3-induced IL-4 production in basophils. Nat Immunol 2008; 10:214-22. [PMID: 19098920 DOI: 10.1038/ni.1686] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 10/31/2008] [Indexed: 01/05/2023]
Abstract
The Fc receptor common gamma-chain (FcRgamma) is a widely expressed adaptor bearing an immunoreceptor tyrosine-based activation motif (ITAM) that transduces activation signals from various immunoreceptors. We show here that basophils lacking FcRgamma developed normally and proliferated efficiently in response to interleukin 3 (IL-3) but were very impaired in IL-3-induced production of IL-4 and in supporting T helper type 2 differentiation. Through its transmembrane portion, FcRgamma associated constitutively with the common beta-chain of the IL-3 receptor and signaled by recruiting the kinase Syk. Retrovirus-mediated complementation demonstrated the essential function of the ITAM of FcRgamma in IL-3 signal transduction. Our results identify a previously unknown mechanism whereby FcRgamma functions to 'route' selective cytokine-triggered signals into the ITAM-mediated IL-4 production pathway.
Collapse
|
39
|
Kuroda S, Nishio M, Sasaki T, Horie Y, Kawahara K, Sasaki M, Natsui M, Matozaki T, Tezuka H, Ohteki T, Förster I, Mak TW, Nakano T, Suzuki A. Effective clearance of intracellular Leishmania major in vivo requires Pten in macrophages. Eur J Immunol 2008; 38:1331-40. [PMID: 18398930 DOI: 10.1002/eji.200737302] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Leishmaniases are a major international public health problem, and macrophages are crucial for host resistance to this parasite. To determine if phosphatase and tensin homologue deleted on chromosome ten (Pten), a negative regulator of the PI3K pathway, plays a role in macrophage-mediated resistance to Leishmania, we generated C57BL/6 mice lacking Pten specifically in macrophages (LysMCrePten(flox/flox) mice). Examination of lesions resulting from Leishmania major infection showed that LysMCrePten(flox/flox) mice were more susceptible to the parasite than wild-type (WT) mice in the early phase of the infection, but were eventually able to eliminate the pathogen. In vitro Pten-deficient macrophages showed a reduced ability to kill parasites in response to IFN-gamma treatment, possibly because the mutant cells exhibited decreased TNF secretion that correlated with reductions in inducible nitric oxide synthase expression and nitric oxide production. In response to various TLR ligands, Pten-deficient macrophages produced less TNF and IL-12 but more IL-10 than WT cells. However, analysis of cells in the lymph nodes draining L. major inoculation sites indicated that both LysMCrePten(flox/flox) and WT mice developed normal Th1 responses following L. major infection, in line with the ability of LysMCrePten(flox/flox) mice to eventually eliminate the parasite. Our results indicate that the efficient clearance of intracellular parasites requires Pten in macrophages.
Collapse
Affiliation(s)
- Shoko Kuroda
- Department of Molecular Biology, Akita University Graduate School of Medicine, Akita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bogdan C. Mechanisms and consequences of persistence of intracellular pathogens: leishmaniasis as an example. Cell Microbiol 2008; 10:1221-34. [DOI: 10.1111/j.1462-5822.2008.01146.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
41
|
Anderson CF, Lira R, Kamhawi S, Belkaid Y, Wynn TA, Sacks D. IL-10 and TGF-β Control the Establishment of Persistent and Transmissible Infections Produced by Leishmania tropica in C57BL/6 Mice. THE JOURNAL OF IMMUNOLOGY 2008; 180:4090-7. [DOI: 10.4049/jimmunol.180.6.4090] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Elshafie AI, Ahlin E, Mathsson L, ElGhazali G, Rönnelid J. Circulating Immune Complexes (IC) and IC-Induced Levels of GM-CSF Are Increased in Sudanese Patients with Acute VisceralLeishmania donovaniInfection Undergoing Sodium Stibogluconate Treatment: Implications for Disease Pathogenesis. THE JOURNAL OF IMMUNOLOGY 2007; 178:5383-9. [PMID: 17404324 DOI: 10.4049/jimmunol.178.8.5383] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection with Leishmania donovani is associated with IL-10 as well as with GM-CSF. Immune complexes (IC) exert important functions by stimulation of monocytes/macrophage-mediated production of pro- and anti-inflammatory cytokines in rheumatic diseases. In this investigation, we have explored IC-induced cytokine production during Leishmania infection. Sera from 43 patients with visceral leishmaniasis (VL), 17 patients with post-kala-azar dermal leishmaniasis, and 20 healthy Sudanese controls were precipitated with polyethylene glycol (PEG). The PEG precipitates were added to serum-free PBMC for 20 h,whereupon supernatant levels of IL-1beta, IL-6, IL-10, IL-1 receptor antagonist protein, TNF-alpha, TNF receptor p75, and GM-CSF were investigated using ELISA. Circulating levels of C1q-binding IC were also measured in the serum samples. PEG precipitates from Leishmania-infected patients induced significantly higher levels of GM-CSF (p = 0.0037) and IL-10 (p < 0.0001), as well as of IL-6 (p < 0.0001) and IL-1 receptor antagonist (p = 0.0238) as compared with PEG precipitates from controls. Patients with acute VL as well as VL patients receiving sodium stibogluconate treatment displayed significantly increased levels of PEG precipitate-induced GM-CSF. The induction of GM-CSF by circulating IC was especially prominent in acute VL patients receiving sodium stibogluconate treatment; ANOVA revealed significant interaction between disease activity and treatment for PEG precipitate-induced levels of GM-CSF (disease activity, p = 0.0006; treatment, p = 0.0005; interaction, p = 0.0046). Parallel associations were determined for C1q-binding immune complexes, but not for any cytokine other than GM-CSF. The importance of IC-induced GM-CSF in leishmaniasis warrants further study.
Collapse
|
43
|
Anderson CF, Oukka M, Kuchroo VJ, Sacks D. CD4(+)CD25(-)Foxp3(-) Th1 cells are the source of IL-10-mediated immune suppression in chronic cutaneous leishmaniasis. ACTA ACUST UNITED AC 2007; 204:285-97. [PMID: 17283207 PMCID: PMC2118728 DOI: 10.1084/jem.20061886] [Citation(s) in RCA: 444] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Nonhealing forms of leishmaniasis in humans are commonly associated with elevated levels of the deactivating cytokine IL-10, and in the mouse, normally chronic infections can be cleared in the absence of IL-10. Using a Leishmania major strain that produces nonhealing dermal lesions in a T helper type 1 (Th1) cell–polarized setting, we have analyzed the cellular sources of IL-10 and their relative contribution to immune suppression. IL-10 was produced by innate cells, as well as CD4+CD25+Foxp3+ and CD4+CD25−Foxp3− T cells in the chronic lesion. Nonetheless, only IL-10 production by antigen-specific CD4+CD25−Foxp3− T cells, the majority of which also produced IFN-γ, was necessary for suppression of acquired immunity in Rag−/− reconstituted mice. Surprisingly, Rag−/− mice reconstituted with naive CD4+ T cells depleted of natural T regulatory cells developed more severe infections, associated with elevated levels of IL-10 and, especially, Th2 cytokines in the site. The data demonstrate that IL-10–producing Th1 cells, activated early in a strong inflammatory setting as a mechanism of feedback control, are the principal mediators of T cell–derived IL-10–dependent immune suppression in a chronic intracellular infection.
Collapse
Affiliation(s)
- Charles F Anderson
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
44
|
Lodge R, Descoteaux A. Phagocytosis of Leishmania donovani amastigotes is Rac1 dependent and occurs in the absence of NADPH oxidase activation. Eur J Immunol 2006; 36:2735-44. [PMID: 16955522 DOI: 10.1002/eji.200636089] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Macrophages produce little superoxide during phagocytosis of Leishmania donovani amastigotes. In this study, we characterized molecular events associated with L. donovani amastigotes uptake by mouse macrophages, to further define the mechanisms by which they are internalized without triggering superoxide production. Using transient transfections, we first showed that internalization of L. donovani amastigotes is mediated by the GTPases Rac1 and Arf6, of which Rac1 is recruited and retained on parasite-containing phagosomes. Next, we showed that, whereas internalization of amastigotes induced no superoxide release, co-internalization of serum-opsonized zymozan and amastigotes resulted in superoxide production. Furthermore, in co-internalization experiments, we detected superoxide production in over 95% of phagosomes containing IgG-opsonized SRBC compared to 5% of amastigote-harboring phagosomes. These results suggest that amastigotes evade the ability of macrophages to produce superoxide during phagocytosis. Consistently, we observed that amastigotes induced barely detectable phosphorylation of the NADPH oxidase component p47phox, leading to a defective phagosomal recruitment of p67phox and p47phox. Finally, we showed that amastigotes disrupt phagosomal lipid raft integrity, potentially interfering with NADPH oxidase assembly. Collectively, our results indicate that internalization of L. donovani amastigotes is a Rac1- and Arf6-dependent process that occurs in the absence of significant NADPH oxidase activation.
Collapse
Affiliation(s)
- Robert Lodge
- INRS-Institut Armand Frappier and Centre for host-parasite interactions, Laval, QC, Canada
| | | |
Collapse
|
45
|
Wanderley JLM, Moreira MEC, Benjamin A, Bonomo AC, Barcinski MA. Mimicry of apoptotic cells by exposing phosphatidylserine participates in the establishment of amastigotes of Leishmania (L) amazonensis in mammalian hosts. THE JOURNAL OF IMMUNOLOGY 2006; 176:1834-9. [PMID: 16424214 DOI: 10.4049/jimmunol.176.3.1834] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Signaling through exposed phosphatidylserine (PS) is fundamental for the TGFbeta1-dependent, noninflammatory phagocytosis of apoptotic cells. This same mechanism operates in the internalization of amastigotes of Leishmania (L) amazonensis (L(L)a) in a process quoted as apoptotic mimicry. Now we show that the host modulates PS exposure by the amastigotes and, as a consequence, BALB/c mice-derived amastigotes expose significantly more PS than those derived from C57BL/6 mice. Due to this difference in the density of surface PS molecules, the former are significantly more infective than the latter, both in vivo, in F1 (BALB/c x C57BL/6) mice, and in vitro, in thioglycollate-derived macrophages from this same mouse strain. PS exposure increases with progression of the lesion and reaches its maximum value in amastigotes obtained at the time point when the lesion in C57BL/6 mice begins to decrease in size and the lesions in BALB/c mice are still growing in size. Synthesis of active TGFbeta1, induction of IL-10 message, and inhibition of NO synthesis correlate with the amount of surface PS displayed by viable (propidium iodide-negative) infective amastigote. Furthermore, we also show that, similar to what happens with apoptotic cells, amastigotes of L(L)a are internalized by macropinocytosis. This mechanism of internalization is consistent with the large phagolysosomes characteristic of L(L)a infection. The intensity of macrophage macropinocytic activity is dependent on the amount of surface PS displayed by the infecting amastigote.
Collapse
Affiliation(s)
- João L M Wanderley
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Cāncer, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
46
|
Woelbing F, Kostka SL, Moelle K, Belkaid Y, Sunderkoetter C, Verbeek S, Waisman A, Nigg AP, Knop J, Udey MC, von Stebut E. Uptake of Leishmania major by dendritic cells is mediated by Fcgamma receptors and facilitates acquisition of protective immunity. J Exp Med 2006; 203:177-88. [PMID: 16418399 PMCID: PMC2118064 DOI: 10.1084/jem.20052288] [Citation(s) in RCA: 164] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Accepted: 12/07/2005] [Indexed: 11/08/2022] Open
Abstract
Uptake of Leishmania major by dendritic cells (DCs) results in activation and interleukin (IL)-12 release. Infected DCs efficiently stimulate CD4- and CD8- T cells and vaccinate against leishmaniasis. In contrast, complement receptor 3-dependent phagocytosis of L. major by macrophages (MPhi) leads exclusively to MHC class II-restricted antigen presentation to primed, but not naive, T cells, and no IL-12 production. Herein, we demonstrate that uptake of L. major by DCs required parasite-reactive immunoglobulin (Ig)G and involved FcgammaRI and FcgammaRIII. In vivo, DC infiltration of L. major-infected skin lesions coincided with the appearance of antibodies in sera. Skin of infected B cell-deficient mice and Fcgamma-/- mice contained fewer parasite-infected DCs in vivo. Infected B cell-deficient mice as well as Fcgamma-/- mice (all on the C57BL/6 background) showed similarly increased disease susceptibility as assessed by lesion volumes and parasite burdens. The B cell-deficient mice displayed impaired T cell priming and dramatically reduced IFN-gamma production, and these deficits were normalized by infection with IgG-opsonized parasites. These data demonstrate that DC and MPhi use different receptors to recognize and ingest L. major with different outcomes, and indicate that B cell-derived, parasite-reactive IgG and DC FcgammaRI and FcgammaRIII are essential for optimal development of protective immunity.
Collapse
Affiliation(s)
- Florian Woelbing
- Department of Dermatology and 2Section for Pathophysiology, First Department of Internal Medicine, Johannes Gutenberg-University, Mainz 55131, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Leishmania major infections induce solid immunity to reinfection. Experimental studies in mice indicate that the CD4+ T cells responsible for this immunity include two populations: parasite-dependent T effector cells and parasite-independent central memory T (Tcm) cells. While there currently is no vaccine for leishmaniasis, the existence of a long-lived population of Tcm cells that does not require the continued presence of live parasites suggests that a vaccine that expands these cells might be efficacious.
Collapse
Affiliation(s)
- Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|