1
|
Abdeladhim M, Teixeira C, Ressner R, Hummer K, Dey R, Gomes R, de Castro W, de Araujo FF, Turiansky GW, Iniguez E, Meneses C, Oliveira F, Aronson N, Lacsina JR, Valenzuela JG, Kamhawi S. Lutzomyia longipalpis salivary proteins elicit human innate and adaptive immune responses detrimental to Leishmania parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640210. [PMID: 40196468 PMCID: PMC11974753 DOI: 10.1101/2025.02.25.640210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Leishmania parasites are transmitted via the bite of infected sand flies, whose saliva modulates host immune responses to promote Leishmania infection, especially in unexposed individuals. For humans in endemic areas, the immune consequences of chronic exposure to sand fly saliva remain poorly understood. We performed a human challenge study with Lutzomyia longipalpis, the primary vector of visceral leishmaniasis in the Americas. Fifteen healthy volunteers were exposed multiple times to uninfected Lu. longipalpis bites over the course of a year. PBMCs collected after several exposures were stimulated ex vivo by recombinant Lu. longipalpis salivary proteins to measure cytokine responses. Two salivary proteins, LJM19 and LJL143, elicited TH1-polarized cytokine responses, but with high co-expression of the TH2 cytokine IL-13. LJM19 also induced higher levels of IL-6 and IL-7, while both LJM19 and LJL143 induced the innate cytokines IL-1β and IFN-α. Importantly, TH1 polarization induced by LJM19 or LJL143 in PBMCs correlated with enhanced killing of Leishmania in co-cultured macrophages. Skin biopsies from two volunteers revealed bite site infiltration with CD4-CD8- T cells. Our data suggest that sand fly exposed individuals demonstrate robust innate and adaptive cellular immune responses to vector salivary proteins that can be co-opted to protect humans against Leishmania infection.
Collapse
Affiliation(s)
- Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Clarissa Teixeira
- Laboratory of Immunoparasitology, Department of Biotechnology, Oswaldo Cruz Foundation, Eusébio, Ceará, Brazil
| | - Roseanne Ressner
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States
| | - Kelly Hummer
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States
| | - Regis Gomes
- Laboratory of Immunoparasitology, Department of Biotechnology, Oswaldo Cruz Foundation, Eusébio, Ceará, Brazil
| | - Waldionê de Castro
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Fernanda Fortes de Araujo
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States
| | - George W. Turiansky
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Naomi Aronson
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
| | - Joshua R. Lacsina
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| |
Collapse
|
2
|
Junior JDES, de Souza JL, da Silva LS, da Silva CC, do Nascimento TA, de Souza MLG, da Cunha AF, Batista JDS, Neto JPDM, Guerra MVDF, Ramasawmy R. A fine mapping of single nucleotide variants and haplotype analysis of IL13 gene in patients with Leishmania guyanensis-cutaneous leishmaniasis and plasma cytokines IL-4, IL-5, and IL-13. Front Immunol 2023; 14:1232488. [PMID: 37908348 PMCID: PMC10613733 DOI: 10.3389/fimmu.2023.1232488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/28/2023] [Indexed: 11/02/2023] Open
Abstract
Introduction Leishmaniasis continues to pose a substantial health burden in 97 countries worldwide. The progression and outcome of Leishmania infection are influenced by various factors, including the cytokine milieu, the skin microbiota at the infection site, the specific Leishmania species involved, the genetic background of the host, and the parasite load. In endemic regions to leishmaniasis, only a fraction of individuals infected actually develops the disease. Overexpression of IL-13 in naturally resistant C57BL/6 mice renders them susceptible to L. major infection. Haplotypes constructed from several single nucleotide variant (SNV) along a chromosome fragment may provide insight into any SNV near the fragment that may be genuinely associated with a phenotype in genetic association studies. Methods We investigated nine SNVs (SNV1rs1881457A>C, SNV2rs1295687C>G, SNV3rs2069744C>T, SNV4rs2069747C>T, SNV5rs20541A>G, SNV6rs1295685A>G, SNV7rs848A>C, SNV8rs2069750G >C, and SNV9rs847T>C) spanning the entire IL13 gene in patients with L. guyanensis cutaneous leishmaniasis (Lg-CL). Results Our analysis did not reveal any significant association between the SNVs and susceptibility/protection against Lg-CL development. However, haplotype analysis, excluding SNV4rs2069747 and SNV8rs2069750 due to low minor allele frequency, revealed that carriers of the haplotype CCCTAAC had a 93% reduced likelihood developing Lg-CL. Similarly, the haplotypes ACCCGCT (ORadj=0.02 [95% CI 0.00-0.07]; p-value, 6.0×10-19) and AGCTAAC (ORadj=0.00[95% CI 0.00-0.00]; p-value 2.7×10-12) appeared to provide protection against the development of Lg-CL. Conversely, carriers of haplotype ACCTGCC have 190% increased likelihood of developing Lg-CL (ORadj=2.9 [95%CI 1.68-5.2]; p-value, 2.5×10-6). Similarly, haplotype ACCCAAT (ORadj=2.7 [95%CI 1.5-4.7]; p-value, 3.2×10-5) and haplotype AGCCGCC are associated with susceptibility to the development of Lg-CL (ORadj=1.7[95%CI 1.04-2.8]; p-value, 0.01). In our investigation, we also found a correlation between the genotypes of rs2069744, rs20541, rs1295685, rs847, and rs848 and plasma IL-5 levels among Lg-Cl patients. Furthermore, rs20541 showed a correlation with plasma IL-13 levels among Lg-Cl patients, while rs2069744 and rs848 showed a correlation with plasma IL-4 levels among the same group. Conclusions Overall, our study identifies three haplotypes of IL13 associated with resistance to disease development and three haplotypes linked to susceptibility. These findings suggest the possibility of a variant outside the gene region that may contribute, in conjunction with other genes, to differences in susceptibility and partially to the pathology.
Collapse
Affiliation(s)
- José do Espírito Santo Junior
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
| | - Josué Lacerda de Souza
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazonia Legal (Rede Bionorte), Universidade do Estado do Amazonas, Manaus, Brazil
| | - Lener Santos da Silva
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazonia Legal (Rede Bionorte), Universidade do Estado do Amazonas, Manaus, Brazil
| | - Cilana Chagas da Silva
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
| | - Tuanny Arruda do Nascimento
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
| | | | | | | | | | - Marcus Vinitius de Farias Guerra
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
| | - Rajendranath Ramasawmy
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazonia Legal (Rede Bionorte), Universidade do Estado do Amazonas, Manaus, Brazil
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Genomic Health Surveillance Network: Optimization of Assistance and Research in The State of Amazonas – REGESAM, Manaus, Amazonas, Brazil
| |
Collapse
|
3
|
Diupotex M, Zamora-Chimal J, Gajón JA, Bonifaz LC, Becker I. CXCR5 and TIM-3 expressions define distinct exhausted T cell subsets in experimental cutaneous infection with Leishmania mexicana. Front Immunol 2023; 14:1231836. [PMID: 37691941 PMCID: PMC10485697 DOI: 10.3389/fimmu.2023.1231836] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
T-cell exhaustion is a key stage in chronic infections since it limits immunopathology, but also hinders the elimination of pathogens. Exhausted T (Tex) cells encompass dynamic subsets, including progenitor cells that sustain long-term immunity through their memory/stem like properties, and terminally-differentiated cells, resembling the so-called Tex cells. The presence of Tex cells in chronic leishmaniasis has been reported in humans and murine models, yet their heterogeneity remains unexplored. Using flow cytometry, we identified Tex cells subtypes based on PD-1, CXCR5 and TIM-3 expressions in draining lymph nodes (dLNs) and lesion sites of C57BL/6 mice infected with L. mexicana at 30-, 60- and 90-days post-infection. We showed that infected mice developed a chronic infection characterized by non-healing lesions with a high parasite load and impaired Th1/Th2 cytokine production. Throughout the infection, PD-1+ cells were observed in dLNs, in addition to an enhanced expression of PD-1 in both CD4+ and CD8+ T lymphocytes. We demonstrated that CD4+ and CD8+ T cells were subdivided into PD-1+CXCR5+TIM-3- (CXCR5+), PD-1+CXCR5+TIM-3+ (CXCR5+TIM-3+), and PD-1+CXCR5-TIM-3+ (TIM-3+) subsets. CXCR5+ Tex cells were detected in dLNs during the whole course of the infection, whereas TIM-3+ cells were predominantly localized in the infection sites at day 90. CXCR5+TIM-3+ cells only increased at 30 and 60 days of infection in dLNs, whereas no increase was observed in the lesions. Phenotypic analysis revealed that CXCR5+ cells expressed significantly higher levels of CCR7 and lower levels of CX3CR1, PD-1, TIM-3, and CD39 compared to the TIM-3+ subset. CXCR5+TIM-3+ cells expressed the highest levels of all exhaustion-associated markers and of CX3CR1. In agreement with a less exhausted phenotype, the frequency of proliferating Ki-67 and IFN-γ expressing cells was significantly higher in the CXCR5+ subset within both CD4+ and CD8+ T cells compared to their respective TIM-3+ subsets, whereas CD8+CXCR5+TIM-3+ and CD8+TIM-3+ subsets showed an enhanced frequency of degranulating CD107a+ cells. In summary, we identified a novel, less-differentiated CXCR5+ Tex subset in experimental cutaneous leishmaniasis caused by L. mexicana. Targeting these cells through immune checkpoint inhibitors such as anti-PD-1 or anti PD-L1 might improve the current treatment for patients with the chronic forms of leishmaniasis.
Collapse
Affiliation(s)
- Mariana Diupotex
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Julián A. Gajón
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Laura C. Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
- Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Ingeborg Becker
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
4
|
Oliveira AS, Aredes-Riguetti LM, Pereira BAS, Alves CR, Souza-Silva F. Degron Pathways and Leishmaniasis: Debating Potential Roles of Leishmania spp. Proteases Activity on Guiding Hosts Immune Response and Their Relevance to the Development of Vaccines. Vaccines (Basel) 2023; 11:1015. [PMID: 37376405 DOI: 10.3390/vaccines11061015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 06/29/2023] Open
Abstract
Degrons are short peptide sequences that signalize target sites for protein degradation by proteases. Herein, we bring forth the discussion on degrons present in proteins related to the immune system of Mus musculus that are potential targets for cysteine and serine proteases of Leishmania spp. and their possible roles on host immune regulation by parasites. The Merops database was used to identify protease substrates and proteases sequence motifs, while MAST/MEME Suite was applied to find degron motifs in murine cytokines (IFN-y, IL-4, IL-5, IL-13, IL-17) and transcription factors (NF-kappaB, STAT-1, AP-1, CREB, and BACH2). STRING tool was used to construct an interaction network for the immune factors and SWISS-MODEL server to generate three-dimensional models of proteins. In silico assays confirm the occurrence of degrons in the selected immune response factors. Further analyses were conducted only in those with resolved three-dimensional structures. The predicted interaction network of degron-containing M. musculus proteins shows the possibility that the specific activity of parasite proteases could interfere with the trend of Th1/Th2 immune responses. Data suggest that degrons may play a role in the immune responses in leishmaniases as targets for parasite proteases activity, directing the degradation of specific immune-related factors.
Collapse
Affiliation(s)
- Adriane Silva Oliveira
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Manguinhos, Rio de Janeiro 21040-360, RJ, Brazil
| | - Lara Mata Aredes-Riguetti
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Manguinhos, Rio de Janeiro 21040-360, RJ, Brazil
| | | | - Carlos Roberto Alves
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Manguinhos, Rio de Janeiro 21040-360, RJ, Brazil
| | - Franklin Souza-Silva
- Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, Manguinhos, Rio de Janeiro 21040-360, RJ, Brazil
- Faculdade de Ciências Biológicas e da Saúde, Universidade Iguaçu, Avenida Abílio Augusto Távora, 2134, Dom Rodrigo, Nova Iguaçu 26260-100, RJ, Brazil
| |
Collapse
|
5
|
Bamorovat M, Sharifi I, Aflatoonian MR, Karamoozian A, Tahmouresi A, Jafarzadeh A, Heshmatkhah A, Sharifi F, Salarkia E, Khaleghi T, Khosravi A, Nooshadokht M, Zarandi MB, Barghi M. Prophylactic effect of cutaneous leishmaniasis against COVID-19: a case-control field assessment. Int J Infect Dis 2022; 122:155-161. [PMID: 34571149 PMCID: PMC8461267 DOI: 10.1016/j.ijid.2021.09.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION We assessed the potential relationship between COVID-19 and laboratory-confirmed cutaneous leishmaniasis (CL)-registered cases with a history of scarring, compared with volunteer participants without history of CL. METHODS This case-control retrospective study was conducted in southeastern Iran with a high anthroponotic cutaneous leishmaniasis (ACL) burden. RESULTS Overall, n=1010 CL cases (n=479 male, n=531 female) were evaluated for infection with SARS-CoV-2. In the CL case group, 2 men and 1 woman (0.3% in total) had a mild form of COVID-19 disease; none were hospitalized or died. In contrast, of n=2020 participants without history of CL, n=57 (2.9%) contracted laboratory-confirmed COVID-19, including mild (66.7%), hospitalized (26.3%), critical (3.5%) and fatal (3.5%). There was a strong negative association between CL infection and COVID-19. The burden of COVID-19 in CL-cured participants significantly reduced the morbidity (odds ratio: 0.12; CI: 0.03-0.30; P <0.001) and mortality (percentile: -4.10, -0.02). CONCLUSION Participants with a history of CL scar had significantly reduced incidence of COVID-19 morbidity and mortality. The cross-protection mediated by CL may retard COVID-19 in endemic countries. However, further longitudinal studies are needed to explore the potential profile and duration of this protection offered by CL against COVID-19.
Collapse
Affiliation(s)
- Mehdi Bamorovat
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mohammad Reza Aflatoonian
- Research Center for Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Karamoozian
- Research Center for Modeling in Health, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Abdollah Jafarzadeh
- Department of Immunology, Kerman University of Medical Sciences, Kerman, Iran
| | - Amireh Heshmatkhah
- Dadbin Health Clinic, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Sharifi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ehsan Salarkia
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Tabandeh Khaleghi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmad Khosravi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nooshadokht
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran; Pathobiology Department, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mehdi Borhani Zarandi
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Barghi
- Department of Biochemistry, Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
6
|
Abstract
Cytokines play crucial roles in commencing and coordinating the organized recruitment and activation of immune cells during infection. These molecular regulators play an important part in deciding the fate of disease outcomes in leishmaniasis, a parasitic disease of tropical and subtropical countries. T helper 1 (Th1) cell-mediated inflammatory cytokines usually play a host-protective role, while T helper 2 (Th2) cell activation produces an anti-inflammatory milieu necessary for parasite survival. It is noteworthy that in such a multifaceted disease, the role played by any particular cytokine cannot be generalized as either beneficial or detrimental. For example, a "host-favorable" cytokine in one form of the disease has been found to be "pathogen friendly" in another form of leishmaniasis. On the other hand, the complex signaling network regulating the production of cytokines is further complicated by the nature of the host as well as the presence of other cytokines in the milieu. The present review focuses on the differential roles played by cytokines and the intricate signaling network responsible for the regulation of such cytokines during infection by different species of Leishmania. While many more studies are required in the future to better understand the role of these molecules in both animal models and patient samples, current studies indicate that these molecules are potential candidates to be targeted for therapy against this deadly disease.
Collapse
|
7
|
Zaatar MT, Simaan Y, Karam MC. Exogenous IL-13 exacerbates Leishmania major infection and abrogates acquired immunity to re-infection. Parasitol Res 2022; 121:2009-2017. [PMID: 35536514 DOI: 10.1007/s00436-022-07539-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
Abstract
Cutaneous leishmaniasis is a major global health issue, affecting more than 88 countries with 0.7-1.2 million new cases per year. T helper polarization plays a significant role in disease outcome, with Th1 responses being associated with resistance and Th2 responses being associated with susceptibility. IL-13 is an important Th2 cytokine with structural and functional similarities to IL-4. In this study, we demonstrate that administering exogenous IL-13 to Leishmania major-infected BALB/c mice increases parasite load in the infected paw and decreases tissue levels of the key Th1/Th2 cytokines IFN-γ and IL-4, respectively. Infecting BALB/c mice with a low dose of L. major has previously been shown to confer resistance to re-infection with a higher dose. In this study, we demonstrate that administration of exogenous IL-13 early in the course of the initial low-dose infection abrogates acquired resistance to high-dose re-infection, as measured by infected paw thickness.
Collapse
Affiliation(s)
- Muriel T Zaatar
- Faculty of Arts and Sciences, Biology Department, University of Balamand Dubai, Dubai, United Arab Emirates.
| | - Youssef Simaan
- Faculty of Sciences, Biology Department, University of Balamand, Koura, Lebanon
| | - Marc C Karam
- Faculty of Sciences, Biology Department, University of Balamand, Koura, Lebanon
| |
Collapse
|
8
|
Bamorovat M, Sharifi I, Tavakoli Oliaee R, Jafarzadeh A, Khosravi A. Determinants of Unresponsiveness to Treatment in Cutaneous Leishmaniasis: A Focus on Anthroponotic Form Due to Leishmania tropica. Front Microbiol 2021; 12:638957. [PMID: 34140933 PMCID: PMC8203913 DOI: 10.3389/fmicb.2021.638957] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/19/2021] [Indexed: 12/17/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is a curable disease; however, due to various risk factors, unresponsiveness to CL treatments is inevitable. The treatment of CL has been firmly correlated with multiple determinants, such as demographical, clinical, and environmental factors, the host’s immune response, poor treatment adherence, the parasite’s genetic make-up, and Leishmania RNA virus. This study primarily focuses on the risk factors associated with different therapeutic outcomes following meglumine antimoniate (MA; Glucantime®) treatment and policy approaches to prevent unresponsiveness in CL patients with a focus on anthroponotic form (ACL). Findings suggest that effective preventive and therapeutic measures should be more vigorously implemented, particularly in endemic areas. Accordingly, extensive training is essential to monitor drug unresponsiveness regularly, especially in tropical regions where the disease is prevalent. Since humans are the fundamental reservoir host of ACL due to L. tropica, prompt detection, early diagnosis, and timely and effective treatment could help control this disease. Furthermore, major challenges and gaps remain: efficacious vaccine, new tools, and expert staff are crucial before CL can be definitively controlled.
Collapse
Affiliation(s)
- Mehdi Bamorovat
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Abdollah Jafarzadeh
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmad Khosravi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
9
|
Krayem I, Lipoldová M. Role of host genetics and cytokines in Leishmania infection. Cytokine 2020; 147:155244. [PMID: 33059974 DOI: 10.1016/j.cyto.2020.155244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/20/2020] [Accepted: 08/08/2020] [Indexed: 12/29/2022]
Abstract
Cytokines and chemokines are important regulators of innate and specific responses in leishmaniasis, a disease that currently affects 12 million people. We overviewed the current information about influences of genetically engineered mouse models of cytokine and chemokine on leishmaniasis. We found that genetic background of the host, parasite species and sub-strain, as well as experimental design often modify effects of genetically engineered cytokine genes. Next we analyzed genes and QTLs (quantitative trait loci) that control response to Leishmania species in mouse in order to establish relationship between genetic control of cytokine expression and organ pathology. These studies revealed a network-like complexity of the combined effects of the multiple functionally diverse QTLs and their individual specificity. Genetic control of organ pathology and systemic immune response overlap only partially. Some QTLs control both organ pathology and systemic immune response, but the effects of genes and loci with the strongest impact on disease are cytokine-independent, whereas several loci modify cytokines levels in serum without influencing organ pathology. Understanding this genetic control might be important in development of vaccines designed to stimulate certain cytokine spectrum.
Collapse
Affiliation(s)
- Imtissal Krayem
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic; Department of Natural Sciences, Faculty of Biomedical Engineering, Czech Technical University in Prague, Sítná 3105, 272 01 Kladno, Czech Republic.
| |
Collapse
|
10
|
Mirzaei A, Maleki M, Masoumi E, Maspi N. A historical review of the role of cytokines involved in leishmaniasis. Cytokine 2020; 145:155297. [PMID: 32972825 DOI: 10.1016/j.cyto.2020.155297] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Leishmaniasis is an infectious disease caused by the Leishmania genus, affecting millions of persons in the world. Despite increased studies, no vaccine has been developed against leishmaniasis, and drug resistance is evolving in some Leishmania species (spp). Innate and acquired immune cells and their associated cytokines interplay together to determine the immune responses related outcomes in leishmaniasis. Interferon (IFN)-γ or macrophage activating factor (MAF) is the first effective lymphokine (LK), with a related function to leishmaniasis, discovered in 1979. This review article discussed the history of cytokines involved in Leishmania infection, and it is the first report demonstrating the involvement in the disease by focusing on cutaneous leishmaniasis. Up to now, the role of many cytokines has been determined and the literature review showed that IL-35 is the latest known cytokine involved in leishmaniasis. This review revealed that the cytokines have pleiotropic effects, depending upon the cytokine environment, generated during the infection and the host genetic background or infecting Leishmania spp. Overall, advances in our knowledge of immune cells and their secreted cytokines, contributing to the protection or pathological process of leishmaniasis may help to reach new approaches for immunotherapy.
Collapse
Affiliation(s)
- Asad Mirzaei
- Department of Parasitology, School of Paramedicine, Ilam University of Medical Sciences, Ilam, Iran; Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Maryam Maleki
- Department of Physiology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Elham Masoumi
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran; Research Committee, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran; Department of Medical Immunology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Nahid Maspi
- Department of Parasitology, School of Paramedicine, Ilam University of Medical Sciences, Ilam, Iran; Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
11
|
Taslimi Y, Agbajogu C, Brynjolfsson SF, Masoudzadeh N, Mashayekhi V, Gharibzadeh S, Östensson M, Nakka SS, Mizbani A, Rafati S, Harandi AM. Profiling inflammatory response in lesions of cutaneous leishmaniasis patients using a non-invasive sampling method combined with a high-throughput protein detection assay. Cytokine 2020; 130:155056. [PMID: 32199248 DOI: 10.1016/j.cyto.2020.155056] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Cutaneous leishmaniasis (CL) is an infection caused by Leishmania (L.) protozoa transmitted through the bite of infected sand fly. Previously, invasive sampling of blood and skin along with low throughput methods were used for determination of inflammatory response in CL patients. AIMS/METHODOLOGY We established a novel approach based on a non-invasive adhesive tape-disc sampling combined with a powerful multiplexing technique called proximity extension assay for profiling 92 inflammatory cytokines, chemokines and surface molecules in the lesions of CL patients infected with L. tropica. Sample collection was done non-invasively by using adhesive tape-discs from lesion and normal skin of 33 L. tropica positive patients. RESULTS Out of 92 inflammatory proteins, the level of 34 proteins was significantly increased in the lesions of CL patients compared to their normal skin. This includes the chemokines CCL2, CCL3, CCL4, CXCL1, CXCL5, CXCL9, CXCL10 and CXCL11, together with the interleukins IL-6, IL-8, IL-18, LIF and OSM. The remaining significantly changed inflammatory proteins include 7 surface molecules and receptors: CD5, CD40, CDCP1, 4E-BP1, TNFRSF9, IL-18R1 and OPG as well as 16 other cytokines and proteins: MMP-1, CSF-1, VEGFA, uPA, EN-RAGE, LAP TGF-β1, HGF, MMP-10, CASP-8, TNFSF14, STAMPB, ADA, TRAIL and ST1A1. Further, 13 proteins showed an increasing trend, albeit not statistically significant, in the CL lesions, including TGF-α, CCL23, MCP-2, IL-12B, CXCL6, IL-24, FGF-19, TNFβ, CD6, TRANCE, IL10, SIR2 and CCL20. CONCLUSION We herein report a novel approach based on a non-invasive sampling method combined with the high-throughput protein assay for profiling inflammatory proteins in CL lesions. Using this approach, we could profile inflammatory proteins in the lesions from CL patients. This new non-invasive approach may have implications for studying skin inflammatory mediators in CL and other skin disorders.
Collapse
Affiliation(s)
- Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Christopher Agbajogu
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | | | - Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Vahid Mashayekhi
- Cutaneous Leishmaniasis Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Safoora Gharibzadeh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Malin Östensson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Sravya Sowdamini Nakka
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | | | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran.
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden; Vaccine Evaluation Center, BC Children's Hospital Research Institute, The University of British Columbia, Canada.
| |
Collapse
|
12
|
Ford J, Hughson A, Lim K, Bardina SV, Lu W, Charo IF, Lim JK, Fowell DJ. CCL7 Is a Negative Regulator of Cutaneous Inflammation Following Leishmania major Infection. Front Immunol 2019; 9:3063. [PMID: 30671055 PMCID: PMC6331479 DOI: 10.3389/fimmu.2018.03063] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022] Open
Abstract
The chemokine CCL7 (MCP3) is known to promote the recruitment of many innate immune cell types including monocytes and neutrophils to sites of bacterial and viral infection and eosinophils and basophils to sites of allergic inflammation. CCL7 upregulation has been associated with many inflammatory settings including infection, cardiovascular disease, and the tumor microenvironment. CCL7's pleotropic effects are due in part to its ability to bind numerous chemokine receptors, namely CCR1, CCR2, CCR3, CCR5, and CCR10. CCL7-blockade or CCL7-deficiency is often marked by decreased inflammation and poor pathogen control. In the context of Leishmania major infection, CCL7 is specifically upregulated in the skin one-2 weeks after infection but its role in L. major control is unclear. To determine CCL7's impact on the response to L. major we infected WT and CCL7-/- C57BL/6 mice. L. major infection of CCL7-deficient mice led to an unexpected increase in inflammation in the infected skin 2 weeks post-infection. A broad increase in immune cell subsets was observed but was dominated by enhanced neutrophilic infiltration. Increased neutrophil recruitment was associated with an enhanced IL-17 gene profile in the infected skin. CCL7 was shown to directly antagonize neutrophil migration in vitro and CCL7 add-back in vivo specifically reduced neutrophil influx into the infected skin revealing an unexpected role for CCL7 in limiting neutrophil recruitment during L. major infection. Enhanced neutrophilic infiltration in CCL7-deficient mice changed the balance of L. major infected host cells with an increase in the ratio of infected neutrophils over monocytes/macrophages. To determine the consequence of CCL7 deficiency on L. major control we analyzed parasite load cutaneously at the site of infection and viscerally in the draining LN and spleen. The CCL7-/- mice supported robust cutaneous parasite control similar to their WT C57BL/6 counterparts. In contrast, CCL7-deficiency led to greater parasite dissemination and poor parasite control in the spleen. Our studies reveal a novel role for CCL7 in negatively regulating cutaneous inflammation, specifically neutrophils, early during L. major infection. We propose that CCL7-mediated dampening of the early immune response in the skin may limit the ability of the parasite to disseminate without compromising cutaneous control.
Collapse
Affiliation(s)
- Jill Ford
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, United States
| | - Angela Hughson
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, United States
| | - Kihong Lim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, United States
| | - Susana V Bardina
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Wuyuan Lu
- Department of Biochemistry and Molecular Biology, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Israel F Charo
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Deborah J Fowell
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, United States
| |
Collapse
|
13
|
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
|
14
|
Abad Dar M, Hölscher C. Arginase-1 Is Responsible for IL-13-Mediated Susceptibility to Trypanosoma cruzi Infection. Front Immunol 2018; 9:2790. [PMID: 30555475 PMCID: PMC6281981 DOI: 10.3389/fimmu.2018.02790] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 11/13/2018] [Indexed: 01/17/2023] Open
Abstract
Arginase-1 (Arg-1) is a marker for alternatively activated macrophages (AAM) and is mainly induced by the type 2 cytokines interleukin (IL)-4 and IL-13 through the common IL-4 receptor-alpha (Rα) subunit. Both, Arg-1 and AAM undermine macrophage effector functions against intracellular parasites and are therefore implicated in the susceptibility to infection with Trypanosoma cruzi, the causative agent of Chagas' disease. However, the involvement of Arg-1 in promoting intracellular replication of T. cruzi in AAM has not been proven so far in vivo. Because Arg-1 is only moderately expressed in T. cruzi-infected wildtype mice, we elucidated the role of Arg-1 and AAM during infection in IL-13-overexpressing (IL-13tg) mice, which are characterized by an inflammation-induced development of AAM and an accompanied elevated expression of Arg-1. In comparison to wildtype littermates, IL-13tg mice were highly susceptible to T. cruzi infection with enhanced parasitemia and impaired survival. Importantly, T. cruzi-infected IL-13tg mice developed an elevated alternative macrophage activation with increased arginase activity. To proof the hypothesis, that Arg-1 accounts for the increased susceptibility of IL-13tg mice, we blocked arginase activity in infected IL-13tg mice. Because this arginase inhibition resulted in a decreased susceptibility to experimental Chagas disease our study supports in summary the conclusion that IL-13/IL-4Rα-driven Arg-1 expression contributes to the permissiveness of the host to T. cruzi infection.
Collapse
Affiliation(s)
- Mahin Abad Dar
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | | |
Collapse
|
15
|
Deletion of Interleukin-4 Receptor Alpha-Responsive Keratinocytes in BALB/c Mice Does Not Alter Susceptibility to Cutaneous Leishmaniasis. Infect Immun 2018; 86:IAI.00710-18. [PMID: 30275010 PMCID: PMC6246911 DOI: 10.1128/iai.00710-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/24/2018] [Indexed: 11/25/2022] Open
Abstract
The skin microenvironment at the site of infection plays a role in the early events that determine protective T helper 1/type 1 immune responses during cutaneous leishmaniasis (CL) infection. During CL in nonhealing BALB/c mice, early interleukin-4 (IL-4) can instruct dendritic cells for protective Th1 immunity. The skin microenvironment at the site of infection plays a role in the early events that determine protective T helper 1/type 1 immune responses during cutaneous leishmaniasis (CL) infection. During CL in nonhealing BALB/c mice, early interleukin-4 (IL-4) can instruct dendritic cells for protective Th1 immunity. Additionally, keratinocytes, which are the principal cell type in the skin epidermis, have been shown to secrete IL-4 early after Leishmania major infection. Here, we investigated whether IL-4/IL-13 signaling via the common IL-4 receptor alpha chain (IL-4Rα) on keratinocytes contributes to susceptibility during experimental CL. To address this, keratinocyte-specific IL-4Rα-deficient (KRT14cre IL-4Rα−/lox) mice on a BALB/c genetic background were generated by gene targeting and site-specific recombination (Cre/loxP) under the control of the keratinocyte-specific krt14 locus. Following high-dose infection with L. major IL-81 and LV39 promastigotes subcutaneously in the footpad, footpad swelling, parasite burden, IFN-γ/IL-4/IL-13 cytokine production, and type 1 and type 2 antibody responses were similar between KRT14cre IL-4Rα−/lox and littermate control IL-4Rα−/lox BALB/c mice. An intradermal infection with low-dose L. major IL-81 and LV39 promastigotes in the ear showed results in infected KRT14cre IL-4Rα−/lox BALB/c mice similar to those of littermate control IL-4Rα−/lox BALB/c mice, with the exception of a significant decrease observed in parasite burden only at the site of LV39 infection in the ear. Collectively, our results show that autocrine and paracrine signaling of IL-4/IL-13 through the IL-4Rα chain on keratinocytes does not influence the establishment of a nonhealing Th2 immune response in BALB/c mice during L. major infection.
Collapse
|
16
|
Insulin-Like Growth Factor-I as an Effector Element of the Cytokine IL-4 in the Development of a Leishmania major Infection. Mediators Inflamm 2018; 2018:9787128. [PMID: 30150896 PMCID: PMC6087592 DOI: 10.1155/2018/9787128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 05/11/2018] [Accepted: 07/08/2018] [Indexed: 01/07/2023] Open
Abstract
Certain cytokines modulate the expression of insulin-like growth factor- (IGF-) I. Since IL-4 and IGF-I promote growth of the protozoan Leishmania major, we here addressed their interaction in downregulating the expression of Igf-I mRNA using small interfering RNA (siRNA) in Leishmania major-infected macrophages. Parasitism was decreased in the siRNA-treated cells compared with the nontreated cells, reversed by the addition of recombinant IGF-I (rIGF-I). In IL-4-stimulated macrophages, parasitism and the Igf-I mRNA amount were increased, and the effects were nullified upon siRNA transfection. IGF-I downregulation inhibited both parasite and macrophage arginase activation even in IL-4-stimulated cells. Searching for intracellular signaling components shared by IL-4 and IGF-I, upon siRNA transfection, phosphorylated p44, p38, and Akt proteins were decreased, affecting the phosphatidylinositol-3-kinase (PI3K)/Akt pathway. In L. major-infected C57BL6-resistant mice, the preincubation of the parasite with rIGF-I changed the infection profile to be similar to that of susceptible mice. We conclude that IGF-I constitutes an effector element of IL-4 involving the PI3K/Akt pathway during L. major infection.
Collapse
|
17
|
Hurdayal R, Brombacher F. Interleukin-4 Receptor Alpha: From Innate to Adaptive Immunity in Murine Models of Cutaneous Leishmaniasis. Front Immunol 2017; 8:1354. [PMID: 29176972 PMCID: PMC5686050 DOI: 10.3389/fimmu.2017.01354] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022] Open
Abstract
The interleukin (IL)-4 receptor alpha (IL-4Rα), ubiquitously expressed on both innate and adaptive immune cells, controls the signaling of archetypal type 2 immune regulators; IL-4 and IL-13, which elicit their signaling action by the type 1 IL-4Rα/gamma common and/or the type 2 IL-4Rα/IL-13Rα complexes. Global gene-deficient mouse models targeting IL-4, IL-13, or the IL-4Rα chain, followed by the development of conditional mice and generation of important cell-type-specific IL-4Rα-deficient mouse models, were indeed critical to gaining in-depth understanding of detrimental T helper (Th) 2 mechanisms in type 1-controlled diseases. A primary example being cutaneous leishmaniasis, which is caused by the protozoan parasite Leishmania major, among others. The disease is characterized by localized self-healing cutaneous lesions and necrosis for which, currently, not a single vaccine has made it to a stage that can be considered effective. The spectrum of human leishmaniasis belongs to the top 10 infectious diseases according to the World Health Organization. As such, 350 million humans are at risk of infection and disease, with an incidence of 1.5–2 million new cases being reported annually. A major aim of our research is to identify correlates of host protection and evasion, which may aid in vaccine design and therapeutic interventions. In this review, we focus on the immune-regulatory role of the IL-4Rα chain from innate immune responses to the development of beneficial type 1 and detrimental type 2 adaptive immune responses during cutaneous Leishmania infection. We discuss the cell-specific requirements of the IL-4Rα chain on crucial innate immune cells during L. major infection, including, IL-4Rα-responsive skin keratinocytes, macrophages, and neutrophils, as well as dendritic cells (DCs). The latter, contributing to one of the paradigm shifts with respect to the role of IL-4 instructing DCs in vivo, to promote Th1 responses against L. major. Finally, we extend these innate responses and mechanisms to control of adaptive immunity and the effect of IL-4Rα-responsiveness on T and B lymphocytes orchestrating the development of CD4+ Th1/Th2 and B effector 1/B effector 2 B cells in response to L. major infection in the murine host.
Collapse
Affiliation(s)
- Ramona Hurdayal
- Faculty of Health Sciences, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Department of Molecular and Cell Biology Faculty of Science, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Faculty of Health Sciences, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| |
Collapse
|
18
|
Olmos-Ortiz LM, Barajas-Mendiola MA, Barrios-Rodiles M, Castellano LE, Arias-Negrete S, Avila EE, Cuéllar-Mata P. Trichomonas vaginalisexosome-like vesicles modify the cytokine profile and reduce inflammation in parasite-infected mice. Parasite Immunol 2017; 39. [DOI: 10.1111/pim.12426] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/17/2017] [Indexed: 12/27/2022]
Affiliation(s)
- L. M. Olmos-Ortiz
- Departamento de Biología; Universidad de Guanajuato; Guanajuato Mexico
| | | | - M. Barrios-Rodiles
- Center for Systems Biology; Lunenfeld-Tanenbaum Research Institute; Mount Sinai Hospital; Toronto ON Canada
| | - L. E. Castellano
- Departamento de Ingenierías Química, Electrónica y Biomédica; Universidad de Guanajuato; León México
| | - S. Arias-Negrete
- Departamento de Biología; Universidad de Guanajuato; Guanajuato Mexico
| | - E. E. Avila
- Departamento de Biología; Universidad de Guanajuato; Guanajuato Mexico
| | - P. Cuéllar-Mata
- Departamento de Biología; Universidad de Guanajuato; Guanajuato Mexico
| |
Collapse
|
19
|
Pandey RK, Mehrotra S, Sharma S, Gudde RS, Sundar S, Shaha C. Leishmania donovani-Induced Increase in Macrophage Bcl-2 Favors Parasite Survival. Front Immunol 2016; 7:456. [PMID: 27826299 PMCID: PMC5078497 DOI: 10.3389/fimmu.2016.00456] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/11/2016] [Indexed: 12/21/2022] Open
Abstract
Members of the Bcl-2 family are major regulators of apoptosis in mammalian cells, and hence infection-induced perturbations in their expression could result into elimination of the parasites or creation of a niche favoring survival. In this investigation, we uncover a novel role of host Bcl-2 in sustaining Leishmania donovani infection. A rapid twofold increase in Bcl-2 expression occurred in response to parasite challenge. Downregulation of post infection Bcl-2 increase using siRNA or functional inhibition using Bcl-2 small molecule inhibitors interfered with intracellular parasite survival confirming the necessity of elevated Bcl-2 during infection. An increased nitric oxide (NO) response and reduced parasitic burden was observed upon Bcl-2 inhibition, where restitution of the NO response accounted for parasite mortality. Mechanistic insights revealed a major role of elevated Th2 cytokine IL-13 in parasite-induced Bcl-2 expression via the transcription factor STAT-3, where blocking at the level of IL-13 receptor or downstream kinase JAK-2 dampened Bcl-2 induction. Increase in Bcl-2 was orchestrated through Toll like receptor (TLR)-2-MEK-ERK signaling, and changes in TLR-2 levels affected parasite uptake. In a mouse model of visceral leishmaniasis (VL), Bcl-2 inhibitors partially restored the antimicrobial NO response by at least a twofold increase that resulted in significantly reduced parasite burden. Interestingly, monocytes derived from the peripheral blood of six out of nine human VL subjects demonstrated Bcl-2 expression at significantly higher levels, and sera from these patients showed only marginally quantifiable nitrites. Collectively, our study for the first time reveals a pro-parasitic role of host Bcl-2 and the capacity of host-derived IL-13 to modulate NO levels during infection via Bcl-2. Here, we propose Bcl-2 inhibition as a possible therapeutic intervention for VL.
Collapse
Affiliation(s)
- Rajeev Kumar Pandey
- Cell Death and Differentiation Research Laboratory, National Institute of Immunology , New Delhi , India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University , Amritsar , India
| | - Smriti Sharma
- Department of Medicine, Institute of Medical Sciences, Infectious Disease Research Laboratory, Banaras Hindu University , Varanasi , India
| | | | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Infectious Disease Research Laboratory, Banaras Hindu University , Varanasi , India
| | - Chandrima Shaha
- Cell Death and Differentiation Research Laboratory, National Institute of Immunology , New Delhi , India
| |
Collapse
|
20
|
Maspi N, Abdoli A, Ghaffarifar F. Pro- and anti-inflammatory cytokines in cutaneous leishmaniasis: a review. Pathog Glob Health 2016; 110:247-260. [PMID: 27660895 DOI: 10.1080/20477724.2016.1232042] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is caused by different species of the genus Leishmania. Pro- and anti-inflammatory cytokines play different roles in resistance/susceptibility and the immunopathogenesis of Leishmania infection. The balance and dynamic changes in cytokines may control or predict clinical outcome. T helper 1 (Th1) inflammatory cytokines (especially interferon-γ, tumor necrosis factor-α and interleukin-12) are the crucial factors in the initiation of protective immunity against L. major infection, whereas T helper 2 cytokines including IL-5, IL-4, and IL-13 facilitate the persistence of parasites by downregulating the Th1 immune response. On the other hand, aggravation of inflammatory reactions leads to collateral tissue damage and formation of ulcer. For this reason, immunity system such as T regulatory cells produce regulatory cytokines such as transforming growth factor-β and IL-10 to inhibit possible injures caused by increased inflammatory responses in infection site. In this article, we review the role of pro- and anti-inflammatory cytokines in the immunoprotection and immunopathology of CL.
Collapse
Affiliation(s)
- Nahid Maspi
- a Faculty of Medical Sciences, Department of Parasitology , Tarbiat Modares University , Tehran , Iran
| | - Amir Abdoli
- a Faculty of Medical Sciences, Department of Parasitology , Tarbiat Modares University , Tehran , Iran
| | - Fathemeh Ghaffarifar
- a Faculty of Medical Sciences, Department of Parasitology , Tarbiat Modares University , Tehran , Iran
| |
Collapse
|
21
|
Mansueto P, Vitale G, Di Lorenzo G, Rini GB, Mansueto S, Cillari E. Immunopathology of Leishmaniasis: An Update. Int J Immunopathol Pharmacol 2016; 20:435-45. [PMID: 17880757 DOI: 10.1177/039463200702000302] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Leishmaniasis represents a severe, increasing, public health problem. The perspective of its control is highly dependent on research progress, on therapeutic manipulations of the immune system, and on vaccine development. There is a correlation between the clinical outcome of Leishmania infection and the cytokine response profile. While a protective immune response against Leishmania has been clearly identified to be related to the influence of a type-1 response and IFN-γ production, the precise role of T helper (TH) 2 cytokines in non-healing infections requires further exploration. IL-4 and IL-13 (TH2 cytokines) can promote disease progression in cutaneous leishmaniasis, whereas IL-4 would appear to enhance protective type-1 responses in visceral leishmaniasis. Thus, the TH1/TH2 paradigm of resistance/susceptibility to intracellular parasites is probably an oversimplification of a more complicated network of regulatory/counter regulatory interactions. Moreover, the presence of antigen specific regulatory T cell subsets may provide an environment that contributes to the balance between TH1 and TH2 cells. Finally, the involvement of CD8+ T cells has been described, but the modality of their function in this kind of infection has not been so far elucidated.
Collapse
Affiliation(s)
- P Mansueto
- Dipartimento di Medicina Clinica e delle Patologie Emergenti, University of Palermo, Palermo, Italy
| | | | | | | | | | | |
Collapse
|
22
|
Kammoun-Rebai W, Naouar I, Libri V, Albert M, Louzir H, Meddeb-Garnaoui A, Duffy D. Protein biomarkers discriminate Leishmania major-infected and non-infected individuals in areas endemic for cutaneous leishmaniasis. BMC Infect Dis 2016; 16:138. [PMID: 27009263 PMCID: PMC4806467 DOI: 10.1186/s12879-016-1458-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 03/09/2016] [Indexed: 11/23/2022] Open
Abstract
Background A successful host immune response to infection is dependent upon both innate and adaptive immune effector mechanisms. Cutaneous leishmaniasis results in an adaptive Th1 CD4+ T cell response that efficiently clears the parasite, but may also result in scaring. However the role of innate mechanisms during parasite clearance remains less well defined. Methods We examined a unique cohort of individuals, living in a Leishmania major endemic region, that were stratified among 3 distinct clinical groups in a cross-sectional study. Specifically, patients were classified either as healed (n = 17), asymptomatic (23), or naïve to infection (18) based upon the classical Leishmanin Skin Test (LST) and the presence or absence of scars. Utilizing a multiplexed immunoassay approach we characterized the induced cytokine and chemokine response to L. major. Results A subset of innate immune molecules was induced in all groups. By contrast, T cell-associated cytokines were largely induced in exposed groups as compared to L. major-infection naïve individuals. Two exceptions were IL-17A and IL-12p70, induced and not induced, respectively, in naïve individuals. In addition, GM-CSF was more strongly induced in healed patients as compared to the other two groups. Surprisingly an IL-13 response was the best cytokine for classifying previously infected donors. Conclusions Exploratory data analysis, utilizing principle component analysis (PCA), revealed distinct patient clusters of the healed and naïve groups based on the most differentially induced proteins. Asymptomatic previously infected individuals were more difficult to assign to a particular cluster based on these induced proteins. Analysis of these proteins may enable the identification of biomarkers associated with disease, leading to a better understanding of the protective mechanisms of immune response against leishmaniasis. Electronic supplementary material The online version of this article (doi:10.1186/s12879-016-1458-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wafa Kammoun-Rebai
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, Institut Pasteur de Tunis, Tunis, Tunisia.,University of Tunis El Manar, Tunis, 1068, Tunisia
| | - Ikbel Naouar
- University of Tunis El Manar, Tunis, 1068, Tunisia.,Laboratory of Transmission Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Valentina Libri
- Center for Human Immunology, Institut Pasteur, Paris, France
| | - Matthew Albert
- Center for Human Immunology, Institut Pasteur, Paris, France.,Department of Immunology, Laboratory of Dendritic Cell Immunobiology, Institut Pasteur, Paris, France.,Inserm U818, Paris, France
| | - Hechmi Louzir
- University of Tunis El Manar, Tunis, 1068, Tunisia.,Laboratory of Transmission Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Medicine, Tunis, Tunisia
| | - Amel Meddeb-Garnaoui
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Darragh Duffy
- Center for Human Immunology, Institut Pasteur, Paris, France. .,Department of Immunology, Laboratory of Dendritic Cell Immunobiology, Institut Pasteur, Paris, France. .,Inserm U818, Paris, France.
| |
Collapse
|
23
|
Terrazas C, Varikuti S, Kimble J, Moretti E, Boyaka PN, Satoskar AR. IL-17A promotes susceptibility during experimental visceral leishmaniasis caused by Leishmania donovani. FASEB J 2015; 30:1135-43. [PMID: 26581600 DOI: 10.1096/fj.15-277202] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/09/2015] [Indexed: 12/20/2022]
Abstract
Leishmania donovani is an intracellular parasite that infects professional phagocytes and causes visceral leishmaniasis (VL). The immune response during VL has been extensively studied in the context of T-helper (Th)1 and Th2 responses. Immunity against this parasite is dependent on IFN-γ production and subsequent macrophage activation, and the Th2 response promotes granuloma formation. The cytokine IL-17A is associated with neutrophilic inflammation. Depletion of neutrophils during experimental VL results in enhanced parasitic loads. Furthermore, although patients resistant to VL showed enhanced levels of IL-17A in circulation, little is known about the role of IL-17A during VL infection. Here, we used IL-17A-deficient mice and IL-17A reporter mice to address the role of IL-17A during VL. IL-17A(-/-) mice were highly resistant to VL infection, showing decreased parasites in the liver and spleen. This unexpected phenotype was associated with enhanced IFN-γ production by T cells and decreased accumulation of neutrophils and monocytes, resulting in reduced number of granulomas. We also found γδ T and Th17 cells as the main IL-17A(+) cells during VL infection. Our data reveal an unexpected role of IL-17A rendering susceptibility against L. donovani by regulating the IFN-γ response and promoting detrimental inflammation.
Collapse
Affiliation(s)
- Cesar Terrazas
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sanjay Varikuti
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jennifer Kimble
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Ellen Moretti
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Prosper N Boyaka
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Abhay R Satoskar
- *Department of Pathology, Department of Veterinary Biosciences, and Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
24
|
Nascimento M, Albuquerque T, Nascimento A, Caldas I, Do-Valle-Matta M, Souto J, Talvani A, Bahia M, Galvão L, Câmara A, Guedes P. Impairment of Interleukin-17A Expression in Canine Visceral Leishmaniosis is Correlated with Reduced Interferon-γ and Inducible Nitric Oxide Synthase Expression. J Comp Pathol 2015; 153:197-205. [DOI: 10.1016/j.jcpa.2015.10.174] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/23/2015] [Accepted: 10/06/2015] [Indexed: 11/25/2022]
|
25
|
Hurdayal R, Brombacher F. The role of IL-4 and IL-13 in cutaneous Leishmaniasis. Immunol Lett 2014; 161:179-83. [DOI: 10.1016/j.imlet.2013.12.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 12/28/2013] [Indexed: 10/25/2022]
|
26
|
Ashok D, Acha-Orbea H. Timing is everything: dendritic cell subsets in murine Leishmania infection. Trends Parasitol 2014; 30:499-507. [DOI: 10.1016/j.pt.2014.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/07/2014] [Accepted: 08/08/2014] [Indexed: 02/02/2023]
|
27
|
Kedzierski L, Evans KJ. Immune responses during cutaneous and visceral leishmaniasis. Parasitology 2014; 141:1544-1562. [PMID: 25075460 DOI: 10.1017/s003118201400095x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Leishmania are protozoan parasites spread by a sandfly insect vector and causing a spectrum of diseases collectively known as leishmaniasis. The disease is a significant health problem in many parts of the world, resulting in an estimated 1·3 million new cases and 30 000 deaths annually. Current treatment is based on chemotherapy, which is difficult to administer, expensive and becoming ineffective in several endemic regions. To date there is no vaccine against leishmaniasis, although extensive evidence from studies in animal models indicates that solid protection can be achieved upon immunization. This review focuses on immune responses to Leishmania in both cutaneous and visceral forms of the disease, pointing to the complexity of the immune response and to a range of evasive mechanisms utilized by the parasite to bypass those responses. The amalgam of innate and acquired immunity combined with the paucity of data on the human immune response is one of the major problems currently hampering vaccine development and implementation.
Collapse
Affiliation(s)
- Lukasz Kedzierski
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Pde, Parkville 3052, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Krystal J Evans
- Department of Medical Biology, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Pde, Parkville 3052, Victoria, Australia
| |
Collapse
|
28
|
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
|
29
|
Ashok D, Schuster S, Ronet C, Rosa M, Mack V, Lavanchy C, Marraco SF, Fasel N, Murphy KM, Tacchini-Cottier F, Acha-Orbea H. Cross-presenting dendritic cells are required for control of Leishmania major infection. Eur J Immunol 2014; 44:1422-32. [PMID: 24643576 DOI: 10.1002/eji.201344242] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/27/2013] [Accepted: 02/06/2014] [Indexed: 11/08/2022]
Abstract
Leishmania major infection induces self-healing cutaneous lesions in C57BL/6 mice. Both IL-12 and IFN-γ are essential for the control of infection. We infected Jun dimerization protein p21SNFT (Batf3(-/-) ) mice (C57BL/6 background) that lack the major IL-12 producing and cross-presenting CD8α(+) and CD103(+) DC subsets. Batf3(-/-) mice displayed enhanced susceptibility with larger lesions and higher parasite burden. Additionally, cells from draining lymph nodes of infected Batf3(-/-) mice secreted less IFN-γ, but more Th2- and Th17-type cytokines, mirrored by increased serum IgE and Leishmania-specific immunoglobulin 1 (Th2 indicating). Importantly, CD8α(+) DCs isolated from lymph nodes of L. major-infected mice induced significantly more IFN-γ secretion by L. major-stimulated immune T cells than CD103(+) DCs. We next developed CD11c-diptheria toxin receptor: Batf3(-/-) mixed bone marrow chimeras to determine when the DCs are important for the control of infection. Mice depleted of Batf-3-dependent DCs from day 17 or wild-type mice depleted of cross-presenting DCs from 17-19 days after infection maintained significantly larger lesions similar to mice whose Batf-3-dependent DCs were depleted from the onset of infection. Thus, we have identified a crucial role for Batf-3-dependent DCs in protection against L. major.
Collapse
Affiliation(s)
- Devika Ashok
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Deletion of IL-4 receptor alpha on dendritic cells renders BALB/c mice hypersusceptible to Leishmania major infection. PLoS Pathog 2013; 9:e1003699. [PMID: 24204259 PMCID: PMC3812013 DOI: 10.1371/journal.ppat.1003699] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 08/28/2013] [Indexed: 02/05/2023] Open
Abstract
In BALB/c mice, susceptibility to infection with the intracellular parasite Leishmania major is driven largely by the development of T helper 2 (Th2) responses and the production of interleukin (IL)-4 and IL-13, which share a common receptor subunit, the IL-4 receptor alpha chain (IL-4Rα). While IL-4 is the main inducer of Th2 responses, paradoxically, it has been shown that exogenously administered IL-4 can promote dendritic cell (DC) IL-12 production and enhance Th1 development if given early during infection. To further investigate the relevance of biological quantities of IL-4 acting on DCs during in vivo infection, DC specific IL-4Rα deficient (CD11c(cre)IL-4Rα(-/lox)) BALB/c mice were generated by gene targeting and site-specific recombination using the cre/loxP system under control of the cd11c locus. DNA, protein, and functional characterization showed abrogated IL-4Rα expression on dendritic cells and alveolar macrophages in CD11c(cre)IL-4Rα(-/lox) mice. Following infection with L. major, CD11c(cre)IL-4Rα(-/lox) mice became hypersusceptible to disease, presenting earlier and increased footpad swelling, necrosis and parasite burdens, upregulated Th2 cytokine responses and increased type 2 antibody production as well as impaired classical activation of macrophages. Hypersusceptibility in CD11c(cre)IL-4Rα(-/lox) mice was accompanied by a striking increase in parasite burdens in peripheral organs such as the spleen, liver, and even the brain. DCs showed increased parasite loads in CD11c(cre)IL-4Rα(-/lox) mice and reduced iNOS production. IL-4Rα-deficient DCs produced reduced IL-12 but increased IL-10 due to impaired DC instruction, with increased mRNA expression of IL-23p19 and activin A, cytokines previously implicated in promoting Th2 responses. Together, these data demonstrate that abrogation of IL-4Rα signaling on DCs is severely detrimental to the host, leading to rapid disease progression, and increased survival of parasites in infected DCs due to reduced killing effector functions.
Collapse
|
31
|
Okwor I, Mou Z, Liu D, Uzonna J. Protective immunity and vaccination against cutaneous leishmaniasis. Front Immunol 2012; 3:128. [PMID: 22661975 PMCID: PMC3361738 DOI: 10.3389/fimmu.2012.00128] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 05/03/2012] [Indexed: 11/21/2022] Open
Abstract
Although a great deal of knowledge has been gained from studies on the immunobiology of leishmaniasis, there is still no universally acceptable, safe, and effective vaccine against the disease. This strongly suggests that we still do not completely understand the factors that control and/or regulate the development and sustenance of anti-Leishmania immunity, particularly those associated with secondary (memory) immunity. Such an understanding is critically important for designing safe, effective, and universally acceptable vaccine against the disease. Here we review the literature on the correlate of protective anti-Leishmania immunity and vaccination strategies against leishmaniasis with a bias emphasis on experimental cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Ifeoma Okwor
- Department of Medical Microbiology, University of Manitoba Winnipeg, MB, Canada
| | | | | | | |
Collapse
|
32
|
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: 105] [Impact Index Per Article: 8.1] [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
|
33
|
Karmakar S, Bhaumik SK, Paul J, De T. TLR4 and NKT cell synergy in immunotherapy against visceral leishmaniasis. PLoS Pathog 2012; 8:e1002646. [PMID: 22511870 PMCID: PMC3325212 DOI: 10.1371/journal.ppat.1002646] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 03/01/2012] [Indexed: 02/06/2023] Open
Abstract
NKT cells play an important role in autoimmune diseases, tumor surveillance, and infectious diseases, providing in most cases protection against infection. NKT cells are reactive to CD1d presented glycolipid antigens. They can modulate immune responses by promoting the secretion of type 1, type 2, or immune regulatory cytokines. Pathogen-derived signals to dendritic cells mediated via Toll like Receptors (TLR) can be modulated by activated invariant Natural Killer T (iNKT) cells. The terminal β-(1–4)-galactose residues of glycans can modulate host responsiveness in a T helper type-1 direction via IFN-γ and TLRs. We have attempted to develop a defined immunotherapeutic, based on the cooperative action of a TLR ligand and iNKT cell using a mouse model of visceral leishmaniasis. We evaluated the anti-Leishmania immune responses and the protective efficacy of the β-(1–4)-galactose terminal NKT cell ligand glycosphingophospholipid (GSPL) antigen of L. donovani parasites. Our results suggest that TLR4 can function as an upstream sensor for GSPL and provoke intracellular inflammatory signaling necessary for parasite killing. Treatment with GSPL was able to induce a strong effective T cell response that contributed to effective control of acute parasite burden and led to undetectable parasite persistence in the infected animals. These studies for the first time demonstrate the interactions between a TLR ligand and iNKT cell activation in visceral leishmaniasis immunotherapeutic. Kala azar (visceral leishmaniasis) is a deadly disease caused by the parasitic protozoa Leishmania donovani. In absence of a suitable vaccine, the incidence of leishmaniasis has increased. The World Health Organization observes that, if the disease is not treated, the fatality rate in developing countries can be as high as 100% within 2 years. Therapy of visceral leishmaniasis can be complicated by toxic side effects, drug resistance, and the need for prolonged treatment regimens. Therefore, improved therapy for leishmaniasis remains desirable. Immunotherapy to selectively induce type 1 immune responses considered essential for resistance to leishmaniasis has shown great promise. CD1d-binding glycolipids stimulate TCR signaling and activation of invariant natural killer T (iNKT) cells. Terminal β-(1–4)-galactose residues in glycoconjugates have been identified as the TLR ligand that induces IFN-γ via TLR signaling. We have used the β-(1–4)-galactose terminal glycosphingophospholipid (GSPL) antigen from L. donovani parasites to treat infected BALB/c mice. We report that immunotherapy with GSPL induced IFN-γ, a type 1 cytokine, through the cooperative action of TLR4 and NKT-cells that contributed to effective control of acute parasite burden in the infected animals.
Collapse
MESH Headings
- Animals
- Antigen Presentation/drug effects
- Antigen Presentation/genetics
- Antigens, CD1d/genetics
- Antigens, CD1d/immunology
- Antigens, CD1d/metabolism
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Antigens, Protozoan/metabolism
- Antigens, Protozoan/pharmacology
- Cricetinae
- Glycosphingolipids/genetics
- Glycosphingolipids/immunology
- Glycosphingolipids/metabolism
- Glycosphingolipids/pharmacology
- Immunotherapy/methods
- Leishmania donovani/immunology
- Leishmania donovani/metabolism
- Leishmaniasis, Visceral/genetics
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/therapy
- Mice
- Mice, Inbred BALB C
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Polysaccharides/genetics
- Polysaccharides/immunology
- Polysaccharides/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/immunology
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
| | | | | | - Tripti De
- Division of Infectious Disease and Immunology, Indian Institute of Chemical Biology, Council of Scientific and Industrial Research, Kolkata, India
- * E-mail:
| |
Collapse
|
34
|
Redundant Notch1 and Notch2 signaling is necessary for IFNγ secretion by T helper 1 cells during infection with Leishmania major. PLoS Pathog 2012; 8:e1002560. [PMID: 22396647 PMCID: PMC3291656 DOI: 10.1371/journal.ppat.1002560] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 01/17/2012] [Indexed: 12/20/2022] Open
Abstract
The protective immune response to intracellular parasites involves in most cases the differentiation of IFNγ-secreting CD4+ T helper (Th) 1 cells. Notch receptors regulate cell differentiation during development but their implication in the polarization of peripheral CD4+ T helper 1 cells is not well understood. Of the four Notch receptors, only Notch1 (N1) and Notch2 (N2) are expressed on activated CD4+ T cells. To investigate the role of Notch in Th1 cell differentiation following parasite infection, mice with T cell-specific gene ablation of N1, N2 or both (N1N2ΔCD4Cre) were infected with the protozoan parasite Leishmania major. N1N2ΔCD4Cre mice, on the C57BL/6 L. major-resistant genetic background, developed unhealing lesions and uncontrolled parasitemia. Susceptibility correlated with impaired secretion of IFNγ by draining lymph node CD4+ T cells and increased secretion of the IL-5 and IL-13 Th2 cytokines. Mice with single inactivation of N1 or N2 in their T cells were resistant to infection and developed a protective Th1 immune response, showing that CD4+ T cell expression of N1 or N2 is redundant in driving Th1 differentiation. Furthermore, we show that Notch signaling is required for the secretion of IFNγ by Th1 cells. This effect is independent of CSL/RBP-Jκ, the major effector of Notch receptors, since L. major-infected mice with a RBP-Jκ deletion in their T cells were able to develop IFNγ-secreting Th1 cells, kill parasites and heal their lesions. Collectively, we demonstrate here a crucial role for RBP-Jκ-independent Notch signaling in the differentiation of a functional Th1 immune response following L. major infection. Infection with protozoan parasites of Leishmania species results in a spectrum of local or systemic diseases in humans and mammals. Overall, leishmaniasis afflicts around 12 million individuals in 88 countries worldwide. Cutaneous leishmaniasis is the most prevalent form of the disease. In order to better understand the complex molecular pathways leading to protection against the cutaneous form of the disease, we used the Leishmania major mouse model. Most mouse strains control L. major infection due to the development of a Th1 response, leading to secretion of IFNγ by T cells which promotes healing and resistance to reinfection. Notch signaling is a very conserved pathway in the regulation of cell differentiation and cell fate decision. However the contribution of Notch receptors in the response to parasite infection is not clear. In this study, we infected mice that do not express Notch1 and Notch2 receptors on the surface of their T cells. We show that these Notch receptors are key players in the development of a protective Th1 immune response against L. major. These results contribute to the understanding of the mechanisms involved in the development of a protective response against pathogens.
Collapse
|
35
|
Tacchini-Cottier F, Weinkopff T, Launois P. Does T Helper Differentiation Correlate with Resistance or Susceptibility to Infection with L. major? Some Insights From the Murine Model. Front Immunol 2012; 3:32. [PMID: 22566916 PMCID: PMC3342012 DOI: 10.3389/fimmu.2012.00032] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 02/13/2012] [Indexed: 11/13/2022] Open
Abstract
The murine model of Leishmania major infection has been an invaluable tool in understanding T helper differentiation in vivo. The initial evidence for a role of distinct CD4+ T helper subsets in the outcome of infection was first obtained with this experimental model. The development of CD4+ Th1 cells was associated with resolution of the lesion, control of parasite replication, and resistance to re-infection in most of the mouse strains investigated (i.e., C57BL/6). In contrast, differentiation of CD4+ Th2 cells correlated with the development of unhealing lesions, and failure to control parasite load in a few strains (i.e., BALB/c). Since these first reports, an incredible amount of effort has been devoted to understanding the various parameters involved in the differentiation of these, and more recently discovered T helper subsets such as Th17 and T regulatory cells. The discovery of cross-talk between T helper subsets, as well as their plasticity force us to reevaluate the events driving a protective/deleterious T helper immune response following infection with L. major in mice. In this review, we describe the individual contributions of each of these CD4+ T helper subsets following L. major inoculation, emphasizing recent advances in the field, such as the impact of different substrains of L. major on the pathogenesis of disease.
Collapse
Affiliation(s)
- Fabienne Tacchini-Cottier
- Department of Biochemistry, WHO Immunology Research and Training Center, University of Lausanne Epalinges, Switzerland
| | | | | |
Collapse
|
36
|
Felizardo TC, Gaspar-Elsas MI, Lima GM, Abrahamsohn IA. Lack of signaling by IL-4 or by IL-4/IL-13 has more attenuating effects on Leishmania amazonensis dorsal skin – than on footpad-infected mice. Exp Parasitol 2012; 130:48-57. [DOI: 10.1016/j.exppara.2011.09.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 09/27/2011] [Accepted: 09/30/2011] [Indexed: 10/16/2022]
|
37
|
Gene Therapy against Murine Melanoma B16F10-Nex2 Using IL-13Ralpha2-Fc Chimera and Interleukin 12 in Association with a Cyclopalladated Drug. Transl Oncol 2011; 1:110-20. [PMID: 18795121 DOI: 10.1593/tlo.08115] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Revised: 06/20/2008] [Accepted: 06/23/2008] [Indexed: 02/02/2023] Open
Abstract
Interleukin 13 (IL-13) is immunoregulatory in many diseases, including cancer. The protective or suppressive role of CD1-restricted natural killer T cells (NKT cells) in tumor immunosurveillance and immunity is well documented. Interleukin 12 (IL-12) can activate type I NKT cells to produce interferon-gamma (IFN-gamma), whereas type II NKT cells may produce IL-13. The high-affinity chain of IL-13Ralpha2 may act as negative inhibitor, suppressing the action of IL-13 and helping to maintain tumor immunosurveillance. We constructed an mIL-13Ralpha2-Fc chimera in a eukaryotic expression vector and confirmed the identity of the recombinant protein by immunoblot analysis and binding to IL-13 in chemiluminescent ELISA. Such DNA vaccine was tested against syngeneic B16F10-Nex2 murine melanoma. In vivo experiments showed a protective effect mediated by high production of IFN-gamma and down-regulation of anti-inflammatory interleukins mainly by NKT 1.1(+) T cells. Biochemoterapy in vivo with plasmid encoding mIL-13Ralpha2-Fc in association with plasmid encoding IL-12 and the 7A cyclopalladated drug led to a significant reduction in the tumor evolution with 30% tumor-free mice. We conclude that IL-12 gene therapy, followed by continuous administration of IL-13Ralpha2-Fc gene along with 7A-drug has antitumor activity involving the high production of proinflammatory cytokines and low immune suppression, specifically by NK1.1(+)T cells producing IL-13 and IL-10.
Collapse
|
38
|
McFarlane E, Carter KC, McKenzie AN, Kaye PM, Brombacher F, Alexander J. Endogenous IL-13 plays a crucial role in liver granuloma maturation during Leishmania donovani infection, independent of IL-4Rα-responsive macrophages and neutrophils. J Infect Dis 2011; 204:36-43. [PMID: 21628656 PMCID: PMC3105032 DOI: 10.1093/infdis/jir080] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 09/20/2010] [Indexed: 12/21/2022] Open
Abstract
Previous studies comparing interleukin 4 receptor α (IL-4Rα)(-/-) and interleukin 4 (IL-4)(-/-) BALB/c mice have indicated that interleukin 13 (IL-13), whose receptor shares the IL-4Rα subunit with IL-4, plays a protective role during visceral leishmaniasis. We demonstrate that IL-13(-/-) BALB/c mice were less able to control hepatic growth of Leishmania donovani compared with wild-type mice. This correlated with significantly retarded granuloma maturation in IL-13(-/-) mice, defective interferon γ (IFN-γ) production, and elevated IL-4 and interleukin 10 (IL-10) levels. L. donovani-infected IL-13(-/-) mice also responded poorly to sodium stibogluconate-mediated chemotherapy compared with wild-type BALB/c mice. Because murine lymphocytes do not have IL-13 receptors, we examined the ability of macrophage/neutrophil-specific IL-4Rα(-/-) mice to control primary infection with L. donovani and to respond to chemotherapy. Macrophage/neutrophil-specific IL-4Rα(-/-) mice were as resistant to leishmaniasis as wild-type mice, and chemotherapy retained its efficacy. Consequently, in L. donovani infected BALB/c mice, IL-13 promotes hepatic granuloma formation and controls parasite burdens independently of direct effects on macrophages/neutrophils.
Collapse
Affiliation(s)
- Emma McFarlane
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow
| | - Katharine C. Carter
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow
| | | | - Paul M. Kaye
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, United Kingdom
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine, Health Science Faculty, University of Cape Town, South Africa
| | - James Alexander
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow
| |
Collapse
|
39
|
Asquith KL, Horvat JC, Kaiko GE, Carey AJ, Beagley KW, Hansbro PM, Foster PS. Interleukin-13 promotes susceptibility to chlamydial infection of the respiratory and genital tracts. PLoS Pathog 2011; 7:e1001339. [PMID: 21573182 PMCID: PMC3088704 DOI: 10.1371/journal.ppat.1001339] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 04/06/2011] [Indexed: 12/22/2022] Open
Abstract
Chlamydiae are intracellular bacteria that commonly cause infections of the respiratory and genital tracts, which are major clinical problems. Infections are also linked to the aetiology of diseases such as asthma, emphysema and heart disease. The clinical management of infection is problematic and antibiotic resistance is emerging. Increased understanding of immune processes that are involved in both clearance and immunopathology of chlamydial infection is critical for the development of improved treatment strategies. Here, we show that IL-13 was produced in the lungs of mice rapidly after Chlamydia muridarum (Cmu) infection and promoted susceptibility to infection. Wild-type (WT) mice had increased disease severity, bacterial load and associated inflammation compared to IL-13 deficient (-/-) mice as early as 3 days post infection (p.i.). Intratracheal instillation of IL-13 enhanced bacterial load in IL-13-/- mice. There were no differences in early IFN-g and IL-10 expression between WT and IL-13-/- mice and depletion of CD4+ T cells did not affect infection in IL-13-/- mice. Collectively, these data demonstrate a lack of CD4+ T cell involvement and a novel role for IL-13 in innate responses to infection. We also showed that IL-13 deficiency increased macrophage uptake of Cmu in vitro and in vivo. Moreover, the depletion of IL-13 during infection of lung epithelial cells in vitro decreased the percentage of infected cells and reduced bacterial growth. Our results suggest that enhanced IL-13 responses in the airways, such as that found in asthmatics, may promote susceptibility to chlamydial lung infection. Importantly the role of IL-13 in regulating infection was not limited to the lung as we showed that IL-13 also promoted susceptibility to Cmu genital tract infection. Collectively our findings demonstrate that innate IL-13 release promotes infection that results in enhanced inflammation and have broad implications for the treatment of chlamydial infections and IL-13-associated diseases.
Collapse
Affiliation(s)
- Kelly L. Asquith
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Jay C. Horvat
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Gerard E. Kaiko
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Alison J. Carey
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Australia
| | - Kenneth W. Beagley
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Australia
| | - Philip M. Hansbro
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Paul S. Foster
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| |
Collapse
|
40
|
Reinhard K, Huber M, Wostl C, Hellhund A, Toboldt A, Abass E, Casper B, Joeris T, Herr C, Bals R, Steinhoff U, Lohoff M, Visekruna A. c-Rel promotes type 1 and type 17 immune responses during Leishmania major infection. Eur J Immunol 2011; 41:1388-98. [DOI: 10.1002/eji.201041056] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 12/16/2010] [Accepted: 01/26/2011] [Indexed: 12/23/2022]
|
41
|
Interleukin-13 reduces hyperalgesia and the level of interleukin-1β in BALB/c mice infected with Leishmania major with an up-regulation of interleukin-6. J Neuroimmunol 2011; 234:49-54. [PMID: 21402416 DOI: 10.1016/j.jneuroim.2011.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 01/10/2011] [Accepted: 02/06/2011] [Indexed: 12/22/2022]
Abstract
The anti-inflammatory cytokines interleukin-10 (IL-10) and interleukin-13 (IL-13) were shown to reduce hyperalgesia in some models such as rats exposed to UV rays. In addition, IL-10 was also shown to reduce hyperalgesia in high dose of Leishmania major-induced inflammation in BALB/c mice accompanied by a significant decrease in the levels of interleukin-1β (IL-1β) in the paws of infected mice, while no effect on the levels of IL-6 was observed. In this study, we injected BALB/c mice with a high dose of L. major and treated them with IL-13 (15 ng/animal) for twelve days (excluding the weekends) and hyperalgesia was assessed using thermal pain tests. Furthermore, the levels of IL-1β and IL-6 were also assessed at different post-infection days. Our results show that IL-6 and more importantly IL-1β don't play a direct role in the L. major-induced hyperalgesia and that IL-13 induces this hyperalgesia through the down-regulation of IL-1β and another proinflammatory cytokine (most probably TNF-α). Furthermore, our data show that IL-13 leads to the upregulation of the level IL-6 which initially seems to have no direct role in the induced hyperalgesia. Therefore, we suggest that the L. major-induced hyperalgesia is mainly mediated by the cytokine cascade leading to the production of sympathetic amines.
Collapse
|
42
|
Costa DL, Carregaro V, Lima-Júnior DS, Silva NM, Milanezi CM, Cardoso CR, Giudice Â, de Jesus AR, Carvalho EM, Almeida RP, Silva JS. BALB/c mice infected with antimony treatment refractory isolate of Leishmania braziliensis present severe lesions due to IL-4 production. PLoS Negl Trop Dis 2011; 5:e965. [PMID: 21390155 PMCID: PMC3046967 DOI: 10.1371/journal.pntd.0000965] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 01/13/2011] [Indexed: 11/18/2022] Open
Abstract
Background Leishmania braziliensis is the main causative agent of cutaneous leishmaniasis in Brazil. Protection against infection is related to development of Th1 responses, but the mechanisms that mediate susceptibility are still poorly understood. Murine models have been the most important tools in understanding the immunopathogenesis of L. major infection and have shown that Th2 responses favor parasite survival. In contrast, L. braziliensis–infected mice develop strong Th1 responses and easily resolve the infection, thus making the study of factors affecting susceptibility to this parasite difficult. Methodology/Principal Findings Here, we describe an experimental model for the evaluation of the mechanisms mediating susceptibility to L. braziliensis infection. BALB/c mice were inoculated with stationary phase promastigotes of L. braziliensis, isolates LTCP393(R) and LTCP15171(S), which are resistant and susceptible to antimony and nitric oxide (NO), respectively. Mice inoculated with LTCP393(R) presented larger lesions that healed more slowly and contained higher parasite loads than lesions caused by LTCP15171(S). Inflammatory infiltrates in the lesions and production of IFN-γ, TNF-α, IL-10 and TGF-β were similar in mice inoculated with either isolate, indicating that these factors did not contribute to the different disease manifestations observed. In contrast, IL-4 production was strongly increased in LTCP393(R)-inoculated animals and also arginase I (Arg I) expression. Moreover, anti-IL-4 monoclonal antibody (mAb) treatment resulted in decreased lesion thickness and parasite burden in animals inoculated with LTCP393(R), but not in those inoculated with LTCP15171(S). Conclusion/Significance We conclude that the ability of L. braziliensis isolates to induce Th2 responses affects the susceptibility to infection with these isolates and contributes to the increased virulence and severity of disease associated with them. Since these data reflect what happens in human infection, this model could be useful to study the pathogenesis of the L. braziliensis infection, as well as to design new strategies of therapeutic intervention. Leishmaniasis is a neglected disease that affects more than 12 million people worldwide. In Brazil, the cutaneous disease is more prevalent with about 28,000 new cases reported each year, and L. braziliensis is the main causative agent. The interesting data about the infection with this parasite is the wide variety of clinical manifestations that ranges from single ulcerated lesions to mucocutaneous and disseminated disease. However, experimental models to study the infection with this parasite are difficult to develop due to high resistance of most mouse strains to the infection, and the mechanisms underlying the distinct manifestations remain poorly understood. Here, the authors use a mouse experimental model of infection with different L. braziliensis isolates, known to induce diseases with distinct severity in the human hosts, to elucidate immune mechanisms that may be involved in the different manifestations. They showed that distinct parasite isolates may modulate host response, and increased IL-4 production and Arg I expression was related to more severe disease, resulting in longer length of disease with larger lesions and reduced parasite clearance. These findings may be useful in the identification of immunological targets to control L. braziliensis infection and potential clinical markers of disease progression.
Collapse
Affiliation(s)
- Diego L. Costa
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa Carregaro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Djalma S. Lima-Júnior
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Neide M. Silva
- Biomedical Sciences Institute, Federal University of Uberlândia, Uberlândia, Brazil
| | - Cristiane M. Milanezi
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cristina R. Cardoso
- Department of Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Ângela Giudice
- Immunology Service, Professor Edgar Santos Universitary Hospital, Federal University of Bahia, Salvador, Brazil
| | - Amélia R. de Jesus
- Department of Internal Medicine and Pathology, Federal University of Sergipe, Aracajú, Brazil
| | - Edgar M. Carvalho
- Immunology Service, Professor Edgar Santos Universitary Hospital, Federal University of Bahia, Salvador, Brazil
| | - Roque P. Almeida
- Department of Internal Medicine and Pathology, Federal University of Sergipe, Aracajú, Brazil
| | - João S. Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- * E-mail:
| |
Collapse
|
43
|
Fakiola M, Mishra A, Rai M, Singh SP, O'Leary RA, Ball S, Francis RW, Firth MJ, Radford BT, Miller EN, Sundar S, Blackwell JM. Classification and regression tree and spatial analyses reveal geographic heterogeneity in genome wide linkage study of Indian visceral leishmaniasis. PLoS One 2010; 5:e15807. [PMID: 21209823 PMCID: PMC3013125 DOI: 10.1371/journal.pone.0015807] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 11/24/2010] [Indexed: 11/18/2022] Open
Abstract
Background Genome wide linkage studies (GWLS) have provided evidence for loci controlling visceral leishmaniasis on Chromosomes 1p22, 6q27, 22q12 in Sudan and 6q27, 9p21, 17q11-q21 in Brazil. Genome wide studies from the major focus of disease in India have not previously been reported. Methods and Findings We undertook a GWLS in India in which a primary ∼10 cM (515 microsatellites) scan was carried out in 58 multicase pedigrees (74 nuclear families; 176 affected, 353 total individuals) and replication sought in 79 pedigrees (102 nuclear families; 218 affected, 473 total individuals). The primary scan provided evidence (≥2 adjacent markers allele-sharing LOD≥0.59; nominal P≤0.05) for linkage on Chromosomes 2, 5, 6, 7, 8, 10, 11, 20 and X, with peaks at 6p25.3-p24.3 and 8p23.1-p21.3 contributed to largely by 31 Hindu families and at Xq21.1-q26.1 by 27 Muslim families. Refined mapping confirmed linkage across all primary scan families at 2q12.2-q14.1 and 11q13.2-q23.3, but only 11q13.2-q23.3 replicated (combined LOD = 1.59; P = 0.0034). Linkage at 6p25.3-p24.3 and 8p23.1-p21.3, and at Xq21.1-q26.1, was confirmed by refined mapping for primary Hindu and Muslim families, respectively, but only Xq21.1-q26.1 replicated across all Muslim families (combined LOD 1.49; P = 0.0045). STRUCTURE and SMARTPCA did not identify population genetic substructure related to religious group. Classification and regression tree, and spatial interpolation, analyses confirm geographical heterogeneity for linkages at 6p25.3-p24.3, 8p23.1-p21.3 and Xq21.1-q26.1, with specific clusters of families contributing LOD scores of 2.13 (P = 0.0009), 1.75 (P = 0.002) and 1.84 (P = 0.001), respectively. Conclusions GWLS has identified novel loci that show geographical heterogeneity in their influence on susceptibility to VL in India.
Collapse
Affiliation(s)
- Michaela Fakiola
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Anshuman Mishra
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Madhukar Rai
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shri Prakash Singh
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rebecca A. O'Leary
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Australia
| | - Stephen Ball
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Australia
| | - Richard W. Francis
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Australia
| | - Martin J. Firth
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Australia
| | - Ben T. Radford
- Australian Institute of Marine Science, The UWA Oceans Institute, The University of Western Australia, Crawley, Australia
| | - E. Nancy Miller
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Jenefer M. Blackwell
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
44
|
Castilho TM, Goldsmith-Pestana K, Lozano C, Valderrama L, Saravia NG, McMahon-Pratt D. Murine model of chronic L. (Viannia) panamensis infection: role of IL-13 in disease. Eur J Immunol 2010; 40:2816-29. [PMID: 20827674 PMCID: PMC3289133 DOI: 10.1002/eji.201040384] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Leishmania (Viannia) organisms are the most prevalent etiologic agents of human cutaneous leishmaniasis in the Americas. Nevertheless, our knowledge of the immunological mechanisms exploited by L. (Viannia) organisms remains limited and the mechanisms underlying disease are not well understood. Here, we report the development of a BALB/c mouse model of L. (V.) panamensis infection that is able to reproduce chronic disease, with persistent infection and clinically evident lesions for over 1 year. The immune response of the mouse resembles that found for L. (V.) panamensis-infected patients with chronic and recurrent lesions, presenting a mixed Th1/Th2 response with the presence of TNF-α, IFN-γ, IL-10 and IL-13. Using immunodeficient mice, the critical role for IL-13 and/or IL-4Rα in determining susceptibility to chronic infection was evident. With the induction of healing in the immunodeficient mice, increases in IFN-γ and IL-17 were found, concomitant with parasite control and elimination. Specifically, increases in CD4(+) (but not CD8(+)) T cells producing IFN-γ were observed. These results suggest that IL-13 represents an important target for disease control of L. (V.) panamensis infection. This murine model should be useful to further understand the pathology associated with chronic disease and to develop methods for the treatment and prevention of leishmaniasis caused by L. (Viannia) parasites.
Collapse
Affiliation(s)
| | | | - Caterin Lozano
- Centro Internacional de Entrenamiento e Investigaciones Médicas, Cali, Colombia
| | - Liliana Valderrama
- Centro Internacional de Entrenamiento e Investigaciones Médicas, Cali, Colombia
| | - Nancy G. Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas, Cali, Colombia
| | | |
Collapse
|
45
|
Coêlho ZCB, Teixeira MJ, Mota EF, Frutuoso MS, da Silva JS, Barral A, Barral-Netto M, Pompeu MML. In vitro initial immune response against Leishmania amazonensis infection is characterized by an increased production of IL-10 and IL-13. Braz J Infect Dis 2010. [DOI: 10.1016/s1413-8670(10)70096-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
46
|
Díaz YR, Rojas R, Valderrama L, Saravia NG. T-bet, GATA-3, and Foxp3 expression and Th1/Th2 cytokine production in the clinical outcome of human infection with Leishmania (Viannia) species. J Infect Dis 2010; 202:406-15. [PMID: 20583921 PMCID: PMC4850829 DOI: 10.1086/653829] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND T cell differentiation determines susceptibility and resistance to experimental cutaneous leishmaniasis, yet mixed T1/Th2 responses characterize the clinical spectrum of human infection with Leishmania (Viannia) species. MATERIALS AND METHODS To discern the interrelationship of T cell differentiation and outcome of human infection, we examined factors that regulate T cell differentiation and Th1/Th2 cytokine responses in asymptomatic infection, active and historical chronic and recurrent cutaneous leishmaniasis. T-bet, GATA-3, Foxp3, and cytokine gene expression were quantified by real-time polymerase chain reaction and correlated with interleukin 2, interferon gamma, tumor necrosis factor alpha, interleukin 4, interleukin 13, and interleukin 10 secretion during in vitro response to live Leishmania panamensis. RESULTS Higher GATA-3 expression than T-bet expression occurred throughout the 15 days of coculture with promastigotes; however, neither transcription nor secretion of interleukin 4 was detected. A sustained inverse correlation between GATA-3 expression and secretion of proinflammatory cytokines interferon gamma and tumor necrosis factor alpha was observed in asymptomatic infection. In contrast, higher T-bet expression and a higher ratio of T-bet to GATA-3 characterized active recurrent disease. Down-regulation of T-bet and GATA-3 expression and increased interleukin 2 secretion, compared with control subjects, was directly correlated with Foxp3 expression and interleukin 13 secretion in chronic disease. CONCLUSIONS Regulation of the inflammatory response rather than biased Th1/Th2 response distinguished asymptomatic and recalcitrant outcomes of infection with Leishmnania viannia species.
Collapse
Affiliation(s)
- Yira Rosalba Díaz
- Centro Internacional de Entrenamiento e Investigaciones Médicas [CIDEIM], Avenida 1 Norte No. 3-03, A.A. 5390, Cali, Colombia
| | - Ricardo Rojas
- Centro Internacional de Entrenamiento e Investigaciones Médicas [CIDEIM], Avenida 1 Norte No. 3-03, A.A. 5390, Cali, Colombia
| | - Liliana Valderrama
- Centro Internacional de Entrenamiento e Investigaciones Médicas [CIDEIM], Avenida 1 Norte No. 3-03, A.A. 5390, Cali, Colombia
| | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas [CIDEIM], Avenida 1 Norte No. 3-03, A.A. 5390, Cali, Colombia
| |
Collapse
|
47
|
Revaz-Breton M, Ronet C, Ives A, Torre YHL, Masina S, Tacchini-Cottier F, Launois P. The MyD88 protein 88 pathway is differently involved in immune responses induced by distinct substrains of Leishmania major. Eur J Immunol 2010; 40:1697-707. [PMID: 20333623 DOI: 10.1002/eji.200939821] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Host resistance to Leishmania major is highly dependent on the development of a Th1 immune response. The TLR adaptator myeloid differentiation protein 88 (MyD88) has been implicated in the Th1 immune response associated with the resistant phenotype observed in C57BL/6 mice after infection with L. major. To investigate whether the MyD88 pathway is differentially used by distinct substrains of parasites, MyD88(-/-) C57BL/6 mice were infected with two substrains of L. major, namely L. major LV39 and L. major IR75. MyD88(-/-) mice were susceptible to both substrains of L. major, although with different kinetics of infection. The mechanisms involved during the immune response associated with susceptibility of MyD88(-/-) mice to L. major is however, parasite substrain-dependent. Susceptibility of MyD88(-/-) mice infected with L. major IR75 is a consequence of Th2 immune-deviation, whereas susceptibility of MyD88(-/-) mice to infection with L. major LV39 resulted from an impaired Th1 response. Depletion of regulatory T cells (Treg) partially restored IFN-gamma secretion and the Th1 immune response in MyD88(-/-) mice infected with L. major LV39, demonstrating a role of Treg activity in the development of an impaired Th1 response in these mice.
Collapse
|
48
|
Pozzoli U, Fumagalli M, Cagliani R, Comi GP, Bresolin N, Clerici M, Sironi M. The role of protozoa-driven selection in shaping human genetic variability. Trends Genet 2010; 26:95-9. [PMID: 20097439 DOI: 10.1016/j.tig.2009.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Revised: 12/23/2009] [Accepted: 12/23/2009] [Indexed: 01/22/2023]
Abstract
Protozoa exert a strong selective pressure in humans. The selection signatures left by these pathogens can be exploited to identify genetic modulators of infection susceptibility. We show that protozoa diversity in different geographic locations is a good measure of protozoa-driven selective pressure; protozoa diversity captured selection signatures at known malaria resistance loci and identified several selected single nucleotide polymorphisms in immune and hemolytic anemia genes. A genome-wide search enabled us to identify 5180 variants mapping to 1145 genes that are subjected to protozoa-driven selective pressure. We provide a genome-wide estimate of protozoa-driven selective pressure and identify candidate susceptibility genes for protozoa-borne diseases.
Collapse
Affiliation(s)
- Uberto Pozzoli
- Scientific Institute IRCCS E. Medea, Bioinformatic Laboratory, Via don L. Monza 20, 23842 Bosisio Parini (LC), Italy
| | | | | | | | | | | | | |
Collapse
|
49
|
De Trez C, Magez S, Akira S, Ryffel B, Carlier Y, Muraille E. iNOS-producing inflammatory dendritic cells constitute the major infected cell type during the chronic Leishmania major infection phase of C57BL/6 resistant mice. PLoS Pathog 2009; 5:e1000494. [PMID: 19557162 PMCID: PMC2695779 DOI: 10.1371/journal.ppat.1000494] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 05/28/2009] [Indexed: 02/07/2023] Open
Abstract
Leishmania major parasites reside and multiply in late endosomal compartments of host phagocytic cells. Immune control of Leishmania growth absolutely requires expression of inducible Nitric Oxide Synthase (iNOS/NOS2) and subsequent production of NO. Here, we show that CD11b+ CD11c+ Ly-6C+ MHC-II+ cells are the main iNOS-producing cells in the footpad lesion and in the draining lymph node of Leishmania major-infected C57BL/6 mice. These cells are phenotypically similar to iNOS-producing inflammatory DC (iNOS-DC) observed in the mouse models of Listeria monocytogenes and Brucella melitensis infection. The use of DsRed-expressing parasites demonstrated that these iNOS-producing cells are the major infected population in the lesions and the draining lymph nodes. Analysis of various genetically deficient mouse strains revealed the requirement of CCR2 expression for the recruitment of iNOS-DC in the draining lymph nodes, whereas their activation is strongly dependent on CD40, IL-12, IFN-γ and MyD88 molecules with a partial contribution of TNF-α and TLR9. In contrast, STAT-6 deficiency enhanced iNOS-DC recruitment and activation in susceptible BALB/c mice, demonstrating a key role for IL-4 and IL-13 as negative regulators. Taken together, our results suggest that iNOS-DC represent a major class of Th1-regulated effector cell population and constitute the most frequent infected cell type during chronic Leishmania major infection phase of C57BL/6 resistant mice. Leishmania spp. are protozoan parasites infecting a variety of mammals, including humans and mice. Much information has been gleaned from murine models of Leishmania major infection. The control of L. major infection by resistant C57BL/6 mice requires the secretion of type 1 (Th1) cytokines (i.e. IFN-γ) by T cells as well as the expression of inducible nitric oxide synthase (iNOS) by phagocytic cells. Conversely, susceptible BALB/c mice are unable to control infection and develop a type 2 (Th2) immune response characterized by the secretion of IL-4 and IL-13 cytokines. In this study, we showed that the main iNOS-producing cells in the lesion and the draining lymph node are phenotypically similar to iNOS-producing “inflammatory” dendritic cells (DC), which are already described in the mouse models of Listeria monocytogenes and Brucella melitensis infection. Our data also highlighted a strong association between the recruitment and activation of these inflammatory DC and the resistance to L. major infection. In addition, we showed that iNOS production by these inflammatory DC is positively regulated by Th1 response and negatively by Th2 response. Taken together, our results provide new insights into how innate and adaptive immune responses fight L. major infection. A better understanding of the mechanisms regulating inflammatory DC recruitment and activation could lead to new therapeutic strategies against Leishmania infection.
Collapse
Affiliation(s)
- Carl De Trez
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Brussels, Belgium
| | - Stefan Magez
- Department of Molecular and Cellular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University Yamadaoka, Suita City, Osaka, Japan
| | - Bernhard Ryffel
- University of Orleans, Transgenose Institute, CNRS, UMR 6218, Orleans, France
| | - Yves Carlier
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Brussels, Belgium
| | - Eric Muraille
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Brussels, Belgium
- * E-mail:
| |
Collapse
|
50
|
Ajdary S, Riazi-Rad F, Alimohammadian MH, Pakzad SR. Immune response to Leishmania antigen in anthroponotic cutaneous leishmaniasis. J Infect 2009; 59:139-43. [PMID: 19560211 DOI: 10.1016/j.jinf.2009.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 04/05/2009] [Accepted: 05/13/2009] [Indexed: 01/26/2023]
Abstract
BACKGROUND Leishmania (L.) tropica is the causative agent of anthroponotic cutaneous leishmaniasis (ACL) in Iran. The disease often heals within a year; however, the non-healing forms of disease are also known. The immunologic responses to L. major infection have been studied in depth, however little is known about the immune status of L. tropica-infected patients. MATERIALS AND METHODS This study was conducted to evaluate T-cell responses to Leishmania antigen in non-healing patients, patients with acute lesion, and healthy donors. Peripheral blood mononuclear cells (PBMC) were cultured with antigen and lymphoproliferative responses were determined. Cytokine profile including gamma interferon (IFN-gamma), interleukin (IL)-5, and IL-13 in supernatants of stimulated cells was also determined. RESULTS The results showed PBMC from both groups of patients proliferated vigorously in response to Leishmania antigens. The levels of IFN-gamma and IL-13 were comparable between patients with acute lesions and non-healing patients. Non-healing patients had significantly higher median levels of IL-5 than patients with acute lesions. The cells from healthy individuals did not respond to Leishmania antigens. CONCLUSIONS High levels of IFN-gamma, IL-5, and IL-13 in non-healing patients suggest a mixed Th1/Th2 response, whereas patients with acute lesion respond to infection by Th1-type response.
Collapse
Affiliation(s)
- Soheila Ajdary
- Immunology department, Pasteur Institute of Iran, Tehran, Islamic Republic of Iran.
| | | | | | | |
Collapse
|