1
|
Lafleur A, Daffis S, Mowbray C, Arana B. Immunotherapeutic Strategies as Potential Treatment Options for Cutaneous Leishmaniasis. Vaccines (Basel) 2024; 12:1179. [PMID: 39460345 PMCID: PMC11511131 DOI: 10.3390/vaccines12101179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Cutaneous leishmaniasis (CL), caused by protozoan parasites of the Leishmania genus, is prevalent in tropical and subtropical regions, with important morbidity, particularly in low- to middle-income countries. Current systemic treatments, including pentavalent antimonials and miltefosine, are associated with significant toxicity, reduced efficacy, and are frequently ineffective in cases of severe or chronic CL. Immunotherapies leverage the immune system to combat microbial infection and offer a promising adjunct or alternative approach to the current standard of care for CL. However, the heterogeneous clinical presentation of CL, which is dependent on parasite species and host immunity, may require informed clinical intervention with immunotherapies. This review explores the clinical and immunological characteristics of CL, emphasising the current landscape of immunotherapies in in vivo models and clinical studies. Such immune-based interventions aim to modulate immune responses against Leishmania, with additive therapeutic effects enabling the efficacy of lower drug doses and decreasing the associated toxicity. Understanding the mechanisms that underlie immunotherapy for CL provides critical insights into developing safer and more effective treatments for this neglected tropical disease. Identifying suitable therapeutic candidates and establishing their safety and efficacy are essential steps in this process. However, the feasibility and utility of these treatments in resource-limited settings must also be considered, taking into account factors such as cost of production, temperature stability, and overall patient access.
Collapse
Affiliation(s)
- Andrea Lafleur
- Doctoral Training Centre, University of Oxford, Oxford OX1 3NP, UK
| | - Stephane Daffis
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| | - Charles Mowbray
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| | - Byron Arana
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| |
Collapse
|
2
|
Álvarez L, Marín-García PJ, Llobat L. Genetic haplotypes associated with immune response to Leishmania infantum infection in dogs. Vet Res Commun 2023; 47:1675-1685. [PMID: 37059873 DOI: 10.1007/s11259-023-10123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Leishmaniasis is a zoonotic parasitic disease, and the main reservoir of the parasite is the dog, although recent years have seen an increase in other mammalian species. In the Mediterranean region, where it is an endemic disease, it is caused by the species Leishmania infantum. The Ibizan hound, an autochthonous breed of this region, appears to have a genetic resistance to parasitic infection, whereas other canine breeds, such as the Boxer, are susceptible to infection. These differences are related to the differentiated activation of the immune response, with the Ibizan hound activating the Th1 immune response, whereas the Boxer breed triggers the Th2 immune response. Cytokine levels and genomic haplotypes of several genes involved in the immune response were analysed in twenty-eight Ibizan hound (resistant canine breed model) and twenty-four Boxer (susceptible canine breed) without clinical signs in the Mediterranean region. Cytokine levels were analysed by ELISA commercial kits and haplotypes were studied using CanineHD DNA Analysis BeadChip including 165,480 mapped positions. The results show 126 haplotypes associated with differential immune response in dogs. Specifically, haplotypes in IL12RB1, IL6R, CIITA, THEMIS, NOXA1, HEY2, RAB38, SLC35D2, SLC28A3, RASEF and DAPK1 genes are associated with serum levels of IFN-γ, IL-2, IL-8, and IL-18. These results suggest that the resistance or susceptibility to Leishmania infantum infection could be a consequence of haplotypes in several genes related to immune response. Future studies are needed to elucidate the relationship of these haplotypes with immune response and gene expression regulation.
Collapse
Affiliation(s)
- Luis Álvarez
- Departamento Produccion Y Sanidad Animal, Salud Publica Y Ciencia Y Tecnologia de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Pablo-Jesús Marín-García
- Departamento Produccion Y Sanidad Animal, Salud Publica Y Ciencia Y Tecnologia de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Lola Llobat
- Departamento Produccion Y Sanidad Animal, Salud Publica Y Ciencia Y Tecnologia de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain.
| |
Collapse
|
3
|
Ihim SA, Abubakar SD, Zian Z, Sasaki T, Saffarioun M, Maleknia S, Azizi G. Interleukin-18 cytokine in immunity, inflammation, and autoimmunity: Biological role in induction, regulation, and treatment. Front Immunol 2022; 13:919973. [PMID: 36032110 PMCID: PMC9410767 DOI: 10.3389/fimmu.2022.919973] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
Interleukin-18 (IL-18) is a potent pro-inflammatory cytokine involved in host defense against infections and regulates the innate and acquired immune response. IL-18 is produced by both hematopoietic and non-hematopoietic cells, including monocytes, macrophages, keratinocytes and mesenchymal cell. IL-18 could potentially induce inflammatory and cytotoxic immune cell activities leading to autoimmunity. Its elevated levels have been reported in the blood of patients with some immune-related diseases, including rheumatoid arthritis, systemic lupus erythematosus, type I diabetes mellitus, atopic dermatitis, psoriasis, and inflammatory bowel disease. In the present review, we aimed to summarize the biological properties of IL-18 and its pathological role in different autoimmune diseases. We also reported some monoclonal antibodies and drugs targeting IL-18. Most of these monoclonal antibodies and drugs have only produced partial effectiveness or complete ineffectiveness in vitro, in vivo and human studies. The ineffectiveness of these drugs targeting IL-18 may be largely due to the loophole caused by the involvement of other cytokines and proteins in the signaling pathway of many inflammatory diseases besides the involvement of IL-18. Combination drug therapies, that focus on IL-18 inhibition, in addition to other cytokines, are highly recommended to be considered as an important area of research that needs to be explored.
Collapse
Affiliation(s)
- Stella Amarachi Ihim
- Department of Molecular and Cellular Pharmacology, University of Shizuoka, Shizuoka, Japan
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
- Department of Science Laboratory Technology, University of Nigeria, Nsukka, Nigeria
| | - Sharafudeen Dahiru Abubakar
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan
- Department of Medical Laboratory Science, College of Medical Science, Ahmadu Bello University, Zaria, Nigeria
| | - Zeineb Zian
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Takanori Sasaki
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Mohammad Saffarioun
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Shayan Maleknia
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- *Correspondence: Gholamreza Azizi,
| |
Collapse
|
4
|
Osero BO, Aruleba RT, Brombacher F, Hurdayal R. Unravelling the unsolved paradoxes of cytokine families in host resistance and susceptibility to Leishmania infection. Cytokine X 2020; 2:100043. [PMID: 33415318 PMCID: PMC7773805 DOI: 10.1016/j.cytox.2020.100043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/16/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022] Open
Abstract
Leishmaniasis is a neglected disease caused by protozoan parasites of the genus Leishmania. Successful clearance of Leishmania relies on a robust human immune response and various cytokines have been implicated in resistance and susceptibility to Leishmania infection. Accordingly, various immunotherapeutic approaches involving cytokines and cytokine receptors are being considered as novel avenues of treatment given the limited efficacy of current anti-leishmanial drugs. These approaches target canonical T helper (Th)1/Type 1 cytokines as intended mediators of host-protection to infection whilst concomitantly suppressing Th2/Type 2 cytokines and their anticipated disease-promoting roles. However, the use of cytokine and cytokine receptor gene-deficient mice over the years has challenged this simplistic view of Th1/Type 1-mediated resistance and Th2/Type 2-mediated susceptibility. Indeed, contribution to susceptibility vs resistance is only a partial consequence to cytokine action as the overall response is multi-faceted due to the pleiotropic, redundant, antagonistic and synergistic action of cytokines and interactions with immune cells in the diseased state. Notably, while the responses of certain cytokines are selectively host-protective or characteristic disease-enhancers, some ligands exert a response depending on the parasite-species initiating infection. Paradoxically, others play dual or contradictory roles in different Leishmania immunopathologies. Hence, cytokines in disease is an unsolved paradox and a comprehensive knowledge of cytokine interplay is important to guide the development of novel immunotherapeutics against leishmaniasis. In this review, we characterize various cytokine families in persistence and clearance of the Leishmania parasite and particularly elucidate unsolved cytokine puzzles in leishmaniasis based on information acquired from "gain of knowledge by loss of function" studies in cytokine and cytokine receptor gene-deficient mice.
Collapse
Affiliation(s)
- Bernard Ong'ondo Osero
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Raphael Taiwo Aruleba
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Ramona Hurdayal
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| |
Collapse
|
5
|
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
|
6
|
Vecchié A, Bonaventura A, Toldo S, Dagna L, Dinarello CA, Abbate A. IL-18 and infections: Is there a role for targeted therapies? J Cell Physiol 2020; 236:1638-1657. [PMID: 32794180 DOI: 10.1002/jcp.30008] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 08/01/2020] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-18 is a pro-inflammatory cytokine belonging to the IL-1 family, first identified for its interferon-γ-inducing properties. IL-18 regulates both T helper (Th) 1 and Th2 responses. It acts synergistically with IL-12 in the Th1 paradigm, whereas with IL-2 and without IL-12 it can induce Th2 cytokine production from cluster of differentation (CD)4+ T cells, natural killer (NK cells, NKT cells, as well as from Th1 cells. IL-18 also plays a role in the hemophagocytic lymphohistiocytosis, a life-threatening condition characterized by a cytokine storm that can be secondary to infections. IL-18-mediated inflammation was largely studied in animal models of bacterial, viral, parasitic, and fungal infections. These studies highlight the contribution of either IL-18 overproduction by the host or overresponsiveness of the host to IL-18 causing an exaggerated inflammatory burden and leading to tissue injury. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19). The damage in the later phase of the disease appears to be driven by a cytokine storm, including interleukin IL-1 family members and secondary cytokines like IL-6. IL-18 may participate in this hyperinflammation, as it was previously found to be able to cause injury in the lung tissue of infected animals. IL-18 blockade has become an appealing therapeutic target and has been tested in some IL-18-mediated rheumatic diseases and infantile-onset macrophage activation syndrome. Given its role in regulating the immune response to infections, IL-18 blockade might represent a therapeutic option for COVID-19, although further studies are warranted to investigate more in detail the exact role of IL-18 in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Alessandra Vecchié
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Aldo Bonaventura
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia.,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Stefano Toldo
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Charles A Dinarello
- Department of Medicine and Immunology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonio Abbate
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
7
|
Saresella M, Basilico N, Marventano I, Perego F, La Rosa F, Piancone F, Taramelli D, Banks H, Clerici M. Leishmania infantum infection reduces the amyloid β 42-stimulated NLRP3 inflammasome activation. Brain Behav Immun 2020; 88:597-605. [PMID: 32335194 DOI: 10.1016/j.bbi.2020.04.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/09/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
Activation of the NLRP3 inflammasome has been shown to play a major role in the neuroinflammation that accompanies Alzheimer's disease (AD); interventions that down regulate the NLRP3 inflammasome could thus be beneficial in AD. Parasite infections were recently shown to be associated with improved cognitive functions in Apolipoprotein E4 (ApoE4)-expressing members of an Amazonian tribe. We verified in an in vitro model whether Leishmania infantum infection could reduce NLRP3. Results obtained in an initial experimental model in which PBMC were LPS primed and nigericin-stimulated showed that L. infantum infection significantly reduced ASC-speck formation (i.e. intracellular inflammasome proteins assembly), as well as the production of activated caspase 5 and IL-1β, but increased that of activated caspase 1 and IL-18. Moreover, L. infantum infection induced the generation of an anti-inflammatory milieu by suppressing the production of TNFα and increasing that of IL-10. These results were replicated when cells that had been LPS-primed were stimulated with Aβ42 and infected with L. infantum. Results herein indicate that Leishmania infection favors an anti-inflammatory milieu, which includes the down-regulation of NLRP3 inflammasome activation, possibly to facilitate its survival inside host cells. A side effect of Leishmaniasis would be the hampering of neuroinflammation; this could play a protective role against AD development.
Collapse
Affiliation(s)
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy.
| | | | - Federica Perego
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy.
| | | | | | - Donatella Taramelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20100 Milan, Italy.
| | - Helen Banks
- Centre for Research on Health and Social Care Management (Cergas), SDA Bocconi School of Management, Milan 20100, Italy.
| | - Mario Clerici
- IRCCS Fondazione don Carlo Gnocchi, 20148 Milan, Italy; Department of Physiopathology and Transplants, University of Milan, 20122 Milan, Italy.
| |
Collapse
|
8
|
de Carvalho RVH, Zamboni DS. Inflammasome Activation in Response to Intracellular Protozoan Parasites. Trends Parasitol 2020; 36:459-472. [PMID: 32298633 DOI: 10.1016/j.pt.2020.02.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
Inflammasomes are cytosolic complexes that assemble in response to cellular stress or upon sensing microbial molecules, culminating in cytokine processing and an inflammatory form of cell death called pyroptosis. Inflammasomes are usually composed of a sensor molecule, an adaptor protein, and an inflammatory caspase, such as Caspase-1, which cleaves and activates multiple substrates, including Gasdermin-D, pro-IL-1β, and pro-IL-18. Ultimately, inflammasome activation promotes inflammation and restriction of the microbial infection. In recent years, many studies have addressed the role of inflammasomes during fungal, bacterial, viral, and parasitic diseases, revealing sophisticated aspects of the host-pathogen interaction. In this review, we summarize recent advances on inflammasome activation in response to intracellular parasites, including Leishmania spp., Plasmodium spp., Trypanosoma cruzi, and Toxoplasma gondii.
Collapse
Affiliation(s)
- Renan V H de Carvalho
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dario S Zamboni
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
9
|
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
|
10
|
Dayakar A, Chandrasekaran S, Kuchipudi SV, Kalangi SK. Cytokines: Key Determinants of Resistance or Disease Progression in Visceral Leishmaniasis: Opportunities for Novel Diagnostics and Immunotherapy. Front Immunol 2019; 10:670. [PMID: 31024534 PMCID: PMC6459942 DOI: 10.3389/fimmu.2019.00670] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 03/12/2019] [Indexed: 12/31/2022] Open
Abstract
Leishmaniasis is a parasitic disease of humans, highly prevalent in parts of the tropics, subtropics, and southern Europe. The disease mainly occurs in three different clinical forms namely cutaneous, mucocutaneous, and visceral leishmaniasis (VL). The VL affects several internal organs and is the deadliest form of the disease. Epidemiology and clinical manifestations of VL are variable based on the vector, parasite (e.g., species, strains, and antigen diversity), host (e.g., genetic background, nutrition, diversity in antigen presentation and immunity) and the environment (e.g., temperature, humidity, and hygiene). Chemotherapy of VL is limited to a few drugs which is expensive and associated with profound toxicity, and could become ineffective due to the parasites developing resistance. Till date, there are no licensed vaccines for humans against leishmaniasis. Recently, immunotherapy has become an attractive strategy as it is cost-effective, causes limited side-effects and do not suffer from the downside of pathogens developing resistance. Among various immunotherapeutic approaches, cytokines (produced by helper T-lymphocytes) based immunotherapy has received great attention especially for drug refractive cases of human VL. Therefore, a comprehensive knowledge on the molecular interactions of immune cells or components and on cytokines interplay in the host defense or pathogenesis is important to determine appropriate immunotherapies for leishmaniasis. Here, we summarized the current understanding of a wide-spectrum of cytokines and their interaction with immune cells that determine the clinical outcome of leishmaniasis. We have also highlighted opportunities for the development of novel diagnostics and intervention therapies for VL.
Collapse
Affiliation(s)
| | | | - Suresh V Kuchipudi
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Suresh K Kalangi
- Department of Biosciences, School of Sciences, Indrashil University, Mehsana, India
| |
Collapse
|
11
|
Interleukin-18 in Health and Disease. Int J Mol Sci 2019; 20:ijms20030649. [PMID: 30717382 PMCID: PMC6387150 DOI: 10.3390/ijms20030649] [Citation(s) in RCA: 371] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-18 was originally discovered as a factor that enhanced IFN-γ production from anti-CD3-stimulated Th1 cells, especially in the presence of IL-12. Upon stimulation with Ag plus IL-12, naïve T cells develop into IL-18 receptor (IL-18R) expressing Th1 cells, which increase IFN-γ production in response to IL-18 stimulation. Therefore, IL-12 is a commitment factor that induces the development of Th1 cells. In contrast, IL-18 is a proinflammatory cytokine that facilitates type 1 responses. However, IL-18 without IL-12 but with IL-2, stimulates NK cells, CD4+ NKT cells, and established Th1 cells, to produce IL-3, IL-9, and IL-13. Furthermore, together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Therefore, IL-18 is a cytokine that stimulates various cell types and has pleiotropic functions. IL-18 is a member of the IL-1 family of cytokines. IL-18 demonstrates a unique function by binding to a specific receptor expressed on various types of cells. In this review article, we will focus on the unique features of IL-18 in health and disease in experimental animals and humans.
Collapse
|
12
|
Wang X, Gong P, Zhang X, Li S, Lu X, Zhao C, Yu Q, Wei Z, Yang Y, Liu Q, Yang Z, Li J, Zhang X. NLRP3 Inflammasome Participates in Host Response to Neospora caninum Infection. Front Immunol 2018; 9:1791. [PMID: 30105037 PMCID: PMC6077289 DOI: 10.3389/fimmu.2018.01791] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 07/19/2018] [Indexed: 02/01/2023] Open
Abstract
Neospora caninum is an intracellular protozoan parasite closely related to Toxoplasma gondii that mainly infects canids as the definitive host and cattle as the intermediate host, resulting in abortion in cattle and leading to financial losses worldwide. Commercial vaccines or drugs are not available for the prevention and treatment of bovine neosporosis. Knowledge about the hallmarks of the immune response to this infection could form the basis of important prevention strategies. The innate immune system first responds to invading parasite and subsequently initiates the appropriate adaptive immune response against this parasite. Upon infection, activation of host pattern-recognition receptors expressed by immune cells triggers the innate immune response. Toll-like receptors, NOD-like receptors, and C-type lectin receptors play key roles in recognizing protozoan parasite. Therefore, we aimed to explore the role of the NLRP3 inflammasome during the acute period of N. caninum infection. In vitro results showed that N. caninum infection of murine bone marrow-derived macrophages activated the NLRP3 inflammasome, accompanied by the release of IL-1β and IL-18, cleavage of caspase-1, and induction of cell death. K+ efflux induced by N. caninum infection participated in the activation of the inflammasome. Infection of mice deficient in NLRP3, ASC, and caspase-1/11 resulted in decreased production of IL-18 and reduced IFN-γ in serum. Elevated numbers of monocytes/macrophages and neutrophils were found at the initial infection site, but they failed to limit N. caninum replication. These findings suggest that the NLRP3 inflammasome is involved in the host response to N. caninum infection at the acute stage and plays an important role in limiting parasite growth, and it may enhance Th1 response by inducing production of IFN-γ. These findings may help devise protocols for controlling neosporosis.
Collapse
Affiliation(s)
- Xiaocen Wang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Pengtao Gong
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xu Zhang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shan Li
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xiangyun Lu
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Chunyan Zhao
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Qile Yu
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhengkai Wei
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yongjun Yang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Qun Liu
- National Animal Protozoa Laboratory, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhengtao Yang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Jianhua Li
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xichen Zhang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| |
Collapse
|
13
|
Nakanishi K. Unique Action of Interleukin-18 on T Cells and Other Immune Cells. Front Immunol 2018; 9:763. [PMID: 29731751 PMCID: PMC5920033 DOI: 10.3389/fimmu.2018.00763] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/27/2018] [Indexed: 12/26/2022] Open
Abstract
Interleukin (IL)-18 was originally discovered as a factor that enhances interferon (IFN)-γ production by anti-CD3-stimulated Th1 cells, particularly in association with IL-12. IL-12 is a cytokine that induces development of Th1 cells. IL-18 cannot induce Th1 cell development, but has the capacity to activate established Th1 cells to produce IFN-γ in the presence of IL-12. Thus, IL-18 is regarded as a proinflammatory cytokine that facilitates type 1 responses. However, in the absence of IL-12 but presence of IL-2, IL-18 stimulates natural killer cells, NKT cells, and even established Th1 cells to produce IL-3, IL-9, and IL-13. Thus, IL-18 also facilitates type 2 responses. This unique function of IL-18 contributes to infection-associated allergic diseases. Together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Thus, IL-18 also induces innate-type allergic inflammation. IL-18 belongs to the IL-1 family of cytokines, which share similar molecular structures, receptors structures, and signal transduction pathways. Nevertheless, IL-18 shows a unique function by binding to a specific receptor expressed on distinct types of cells. In this review article, I will focus on the unique features of IL-18 in lymphocytes, basophils, and mast cells, particularly in comparison with IL-33.
Collapse
Affiliation(s)
- Kenji Nakanishi
- Department of Immunology, Hyogo College of Medicine, Hyogo, Japan
| |
Collapse
|
14
|
How Inflammasomes Inform Adaptive Immunity. J Mol Biol 2017; 430:217-237. [PMID: 28987733 DOI: 10.1016/j.jmb.2017.09.019] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 02/07/2023]
Abstract
An immune response consists of a finely orchestrated interplay between initial recognition of potential microbial threats by the innate immune system and subsequent licensed adaptive immune neutralization. The initial recognition integrates environmental cues derived from pathogen-associated molecular patterns and cell-intrinsic damage-associated molecular patterns to contextualize the insult and inform a tailored adaptive response via T and B lymphocytes. While there are much data to support the role of transcriptional responses downstream of pattern recognition receptors in informing the adaptive immune response, markedly less attention has been paid to the role of post-translational responses to pathogen-associated molecular pattern and damage-associated molecular pattern recognition by the innate immune system, and how this may influence adaptive immunity. A well-characterized post-translational consequence of pattern recognition receptor signaling is the assembly of a multimeric signaling platform, termed the inflammasome, by members of the nucleotide-binding oligomerization domain (Nod), leucine-rich repeat-containing receptors (NLRs), and pyrin and HIN domain (PYHIN) families. Inflammasomes assemble in response to cytosolic perturbations, such as mitochondrial dysfunction and aberrant ion fluxes in the case of the canonical NLRP3 inflammasome or the presence of bacterial lipopolysaccharides in the case of the non-canonical inflammasome. Assembly of the inflammasome allows for the cleavage and activation of inflammatory caspases. These activated inflammatory caspases in turn cleave pro-form inflammatory cytokines into their mature bioactive species and lead to unconventional protein secretion and lytic cell death. In this review, we discuss evidence for inflammasome-mediated instruction and contextualization of infectious and sterile agents to the adaptive immune system.
Collapse
|
15
|
Demarchi IG, Terron MDS, Thomazella MV, Mota CA, Gazim ZC, Cortez DAG, Aristides SMA, Silveira TGV, Lonardoni MVC. Antileishmanial and immunomodulatory effects of the essential oil from Tetradenia riparia (Hochstetter) Codd. Parasite Immunol 2016; 38:64-77. [PMID: 26615004 DOI: 10.1111/pim.12297] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 11/24/2015] [Indexed: 12/12/2022]
Abstract
Cutaneous leishmaniasis usually presents therapeutic resistance to antimonials, and the existing therapies for leishmaniasis have many adverse effects and toxicity. Natural products may be regarded as possible candidates for alternative leishmaniasis treatment. The plant Tetradenia riparia has shown promise for the treatment of infectious diseases in folk medicine. We evaluated the antileishmanial activity of an essential oil from T. riparia (TrEO) and the modulatory effects of TrEO on cytokine modulation by peritoneal fluid cells that were infected with L. (L.) amazonensis. Peritoneal fluid cells were infected with Leishmania and incubated with TrEO (30 ng/mL) for 3, 6, and 24 h. Cytokines were screened using semi-quantitative reverse-transcription polymerase chain reaction (RT-PCR) and flow cytometry. Antileishmanial activity was evaluated at 24 h by microscopic counting and quantitative PCR (qPCR). TrEO treatment induced the death of 50% of Leishmania amastigotes (indicated by microscopic counting) and 91% of the parasite load (indicated by qPCR). TrEO inhibited some of the most critical cytokines for parasite growth and the establishment of infection, including granulocyte-macrophage colony-stimulating factor, interleukin-4 (IL-4), IL-10, and tumour necrosis factor. The parasite inhibited interferon-γ and IL-12, and TrEO blocked this inhibition, indicating that these cytokines are critical for activating mechanisms associated with the death and elimination of the parasite. These results suggest that TrEO may be an alternative leishmaniasis therapy when considering its antileishmanial and immunomodulatory activity.
Collapse
Affiliation(s)
- Izabel Galhardo Demarchi
- Departamento de Análises Clínicas e Biomedicina, Laboratório de Imunologia Clínica, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Mariana de Souza Terron
- Departamento de Análises Clínicas e Biomedicina, Laboratório de Imunologia Clínica, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Mateus Vailant Thomazella
- Departamento de Análises Clínicas e Biomedicina, Laboratório de Imunologia Clínica, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Camila Alves Mota
- Departamento de Análises Clínicas e Biomedicina, Laboratório de Imunologia Clínica, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Zilda Cristiani Gazim
- Departamento de Farmácia, Laboratório de Química de Produtos Naturais da Universidade Paranaense, Umuarama, Paraná, Brazil
| | | | - Sandra Mara Alessi Aristides
- Departamento de Análises Clínicas e Biomedicina, Laboratório de Imunologia Clínica, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Thaís Gomes Verzignassi Silveira
- Departamento de Análises Clínicas e Biomedicina, Laboratório de Imunologia Clínica, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Maria Valdrinez Campana Lonardoni
- Departamento de Análises Clínicas e Biomedicina, Laboratório de Imunologia Clínica, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| |
Collapse
|
16
|
Gurung P, Kanneganti TD. Immune responses against protozoan parasites: a focus on the emerging role of Nod-like receptors. Cell Mol Life Sci 2016; 73:3035-51. [PMID: 27032699 PMCID: PMC4956549 DOI: 10.1007/s00018-016-2212-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 03/11/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
Abstract
Nod-like receptors (NLRs) have gained attention in recent years because of the ability of some family members to assemble into a multimeric protein complex known as the inflammasome. The role of NLRs and the inflammasome in regulating innate immunity against bacterial pathogens has been well studied. However, recent studies show that NLRs and inflammasomes also play a role during infections caused by protozoan parasites, which pose a significant global health burden. Herein, we review the diseases caused by the most common protozoan parasites in the world and discuss the roles of NLRs and inflammasomes in host immunity against these parasites.
Collapse
Affiliation(s)
- Prajwal Gurung
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-2794, USA.
| |
Collapse
|
17
|
Saberi S, Arjmand R, Soleimanifard S, Khamesipour A, Hosseini SM, Salehi M, Varshosaz J, Palizban AA, Hejazi SH. In vivo/In vitro immune responses to L. major isolates from patients with no clinical response to Glucantime. Adv Biomed Res 2016; 5:126. [PMID: 27563636 PMCID: PMC4976526 DOI: 10.4103/2277-9175.187000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 10/26/2014] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Leishmaniasis is a major health problem in some endemic areas of tropical and subtropical areas of the world. Interleukin-12 (IL-12) and interferon gamma (IFN-γ) are essential cytokines associated with initiation of Th1 response. The main objective of this study was to evaluate of the type of immune response to L. major isolates from patients with no clinical response to antimonite (Glucantime). MATERIALS AND METHODS This experimental study was carried out during 2013-2014. In the current study Leishmania major were isolated from 10 CL patients with a history of at least one course of treatment with Meglumine antimonate (Sb5). The isolates were used to evaluate in vitro and in vivo response to Sb5. J774 murine macrophage cell line was used for in vitro tests and Balb/c mice was used for in vivo studies. IL-12 gene expression was evaluated using Real-time PCR and IFN-γ serum level was quantified using ELISA technique. SPSS (version: 20), analysis of Covariance (ANCOVA) was used for statistical analysis. RESULTS PCR results confirmed that all 10 isolates were L. major. The mean of IL-12 gene expression in vitro, in vivo and IFN-γ serum levels (pg/ml) after 2 and 3 weeks treatment in vivo, increased significantly following the treatment with Glucantime in the two groups of Balb/c mice infected either with patients' isolates or standard L. major. No significant difference was seen between the patients' isolates and standard species. CONCLUSIONS Although the L. major were isolated from patients with active lesion and no clinical response to Glucantime after at least one courses of Glucantime treatment but in vivo and in vitro immune response of L. major isolates showed no difference between the patients' isolates and standard L. major.
Collapse
Affiliation(s)
- Sedigheh Saberi
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, and Department of Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Arjmand
- Medical Parasitology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Simindokht Soleimanifard
- Medical Parasitology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Hosseini
- Bio-Statistics and Epidemiology, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansoor Salehi
- Biology and Genetics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jaleh Varshosaz
- Pharmaceutics, Novel Drug Delivery Systems Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Ali Palizban
- Clinical Biochemistry Department, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Hossein Hejazi
- Medical Parasitology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Parasitology and Mycology, School of Medicine, Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
18
|
Zamboni DS, Lima-Junior DS. Inflammasomes in host response to protozoan parasites. Immunol Rev 2015; 265:156-71. [DOI: 10.1111/imr.12291] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Dario S. Zamboni
- Department of Cell Biology; School of Medicine of Ribeirão Preto; University of São Paulo; Ribeirão Preto Brazil
| | - Djalma S. Lima-Junior
- Department of Cell Biology; School of Medicine of Ribeirão Preto; University of São Paulo; Ribeirão Preto Brazil
| |
Collapse
|
19
|
Abstract
Parasitic diseases are a serious global health concern. Many of the most common and most severe parasitic diseases, including Chagas' disease, leishmaniasis, and schistosomiasis, are also classified as neglected tropical diseases and are comparatively less studied than infectious diseases prevalent in high income nations. The NLRs (nucleotide-binding domain leucine-rich-repeat-containing proteins) are cytosolic proteins known to be involved in pathogen detection and host response. The role of NLRs in the host response to parasitic infection is just beginning to be understood. The NLR proteins NOD1 and NOD2 have been shown to contribute to immune responses during Trypanosoma cruzi infection, Toxoplasma gondii infection, and murine cerebral malaria. The NLRP3 inflammasome is activated by T. cruzi and Leishmania amazonensis but also induces pathology during infection with schistosomes or malaria. Both the NLRP1 and NLRP3 inflammasomes respond to T. gondii infection. The NLRs may play crucial roles in human immune responses during parasitic infection, usually acting as innate immune sensors and driving the inflammatory response against invading parasites. However, this inflammatory response can either kill the invading parasite or be responsible for destructive pathology. Therefore, understanding the role of the NLR proteins will be critical to understanding the host defense against parasites as well as the fine balance between homeostasis and parasitic disease.
Collapse
Affiliation(s)
- Gwendolyn M Clay
- The Interdisciplinary Program in Molecular and Cellular Biology, University of Iowa, 400 EMRB, 500 Newton Rd., Iowa City, IA, 52242, USA
| | | | | |
Collapse
|
20
|
Verardi PH, Legrand FA, Chan KS, Peng Y, Jones LA, Yilma TD. IL-18 expression results in a recombinant vaccinia virus that is highly attenuated and immunogenic. J Interferon Cytokine Res 2014; 34:169-78. [PMID: 24168450 PMCID: PMC3942681 DOI: 10.1089/jir.2013.0052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/19/2013] [Indexed: 01/06/2023] Open
Abstract
Interferon-γ (IFN-γ) is an attenuating factor for vaccinia virus (VACV), decreasing its virulence in vivo by more than a million fold. It is also a highly effective adjuvant when administered at the time of immunization with protein antigens. However, recombinant VACV (rVACV) vaccines expressing IFN-γ do not induce enhanced immune responses. It is possible that the IFN-γ expressed by rVACVs induces both an antiviral state and increased immunological clearance, thus resulting in decreased levels of antigen expression due to reduced viral replication and spread. We conjectured that delaying expression of IFN-γ would result in enhanced production of antigens by rVACVs thus resulting in increased immune responses to foreign antigens. Interleukin (IL)-18, also known as IFN-γ inducing factor, is a cytokine that induces T and NK cells to produce IFN-γ. In this study, we demonstrated that an rVACV expressing bioactive murine IL-18 replicated to low but detectable levels in vivo, unlike an rVACV expressing IFN-γ. Moreover, the rVACV expressing IL-18 was significantly attenuated in both immunocompromised and immunocompetent mice. This attenuation was dependent on IFN-γ, as IL-18 expression failed to attenuate VACV in IFN-γ knock-out mice. Cytotoxic T-cell (CTL) and anamnestic antibody responses were slightly increased in animals vaccinated with the rVACV expressing IL-18. Thus, induction of IFN-γ because of IL-18 expression resulted in an rVACV that replicated to low but detectable levels in vivo, yet elicited slightly better CTL and anamnestic humoral immune responses.
Collapse
Affiliation(s)
- Paulo H. Verardi
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, Connecticut
| | - Fatema A. Legrand
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
| | - Kenneth S. Chan
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
| | - Yue Peng
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
| | - Leslie A. Jones
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
| | - Tilahun D. Yilma
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
- Department of Medical Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, California
| |
Collapse
|
21
|
Fann DYW, Lee SY, Manzanero S, Chunduri P, Sobey CG, Arumugam TV. Pathogenesis of acute stroke and the role of inflammasomes. Ageing Res Rev 2013; 12:941-66. [PMID: 24103368 DOI: 10.1016/j.arr.2013.09.004] [Citation(s) in RCA: 270] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 09/12/2013] [Accepted: 09/19/2013] [Indexed: 12/20/2022]
Abstract
Inflammation is an innate immune response to infection or tissue damage that is designed to limit harm to the host, but contributes significantly to ischemic brain injury following stroke. The inflammatory response is initiated by the detection of acute damage via extracellular and intracellular pattern recognition receptors, which respond to conserved microbial structures, termed pathogen-associated molecular patterns or host-derived danger signals termed damage-associated molecular patterns. Multi-protein complexes known as inflammasomes (e.g. containing NLRP1, NLRP2, NLRP3, NLRP6, NLRP7, NLRP12, NLRC4, AIM2 and/or Pyrin), then process these signals to trigger an effector response. Briefly, signaling through NLRP1 and NLRP3 inflammasomes produces cleaved caspase-1, which cleaves both pro-IL-1β and pro-IL-18 into their biologically active mature pro-inflammatory cytokines that are released into the extracellular environment. This review will describe the molecular structure, cellular signaling pathways and current evidence for inflammasome activation following cerebral ischemia, and the potential for future treatments for stroke that may involve targeting inflammasome formation or its products in the ischemic brain.
Collapse
|
22
|
Kinoshita M, Miyazaki H, Ono S, Seki S. Immunoenhancing therapy with interleukin-18 against bacterial infection in immunocompromised hosts after severe surgical stress. J Leukoc Biol 2013; 93:689-98. [DOI: 10.1189/jlb.1012502] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
23
|
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
|
24
|
Rasouli M, Kalani M, Moravej A, Kiany S. Interleukin-18 single nucleotide polymorphisms contribute to the susceptibility to brucellosis in Iranian patients. Cytokine 2011; 54:272-6. [DOI: 10.1016/j.cyto.2011.02.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 01/14/2011] [Accepted: 02/09/2011] [Indexed: 11/17/2022]
|
25
|
Doi T, Sakoda T, Akagami T, Naka T, Mori Y, Tsujino T, Masuyama T, Ohyanagi M. Aldosterone induces interleukin-18 through endothelin-1, angiotensin II, Rho/Rho-kinase, and PPARs in cardiomyocytes. Am J Physiol Heart Circ Physiol 2008; 295:H1279-H1287. [PMID: 18660453 DOI: 10.1152/ajpheart.00148.2008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Aldosterone (Aldo) is recognized as an important risk factor for cardiovascular diseases. IL-18 induces myocardial hypertrophy, loss of contractility of cardiomyocytes, and apoptosis leading myocardial dysfunction. However, so far, there have been few reports concerning the interaction between Aldo and IL-18. The present study examined the effects and mechanisms of Aldo on IL-18 expression and the roles of peroxisome proliferator-activated receptor (PPAR) agonists in rat cardiomyocytes. We used cultured rat neonatal cardiomyocytes stimulated with Aldo to measure IL-18 mRNA and protein expression, Rho-kinase, and NF-kappaB activity. We also investigated the effects of PPAR agonists on these actions. Aldo, endothelin-1 (ET-1), and angiotensin II (ANG II) increased IL-18 mRNA and protein expression. Mineralocorticoid receptor antagonists, endothelin A receptor antagonist, and ANG II receptor antagonist inhibited Aldo-induced IL-18 expression. Aldo induced ET-1 and ANG II production in cultured media. Moreover, Rho/Rho-kinase inhibitor and statin inhibited Aldo-induced IL-18 expression. On the other hand, Aldo upregulated the activities of Rho-kinase and NF-kappaB. PPAR agonists attenuated the Aldo-induced IL-18 expression and NF-kappaB activity but not the Rho-kinase activity. Our findings indicate that Aldo induces IL-18 expression through a mechanism that involves, at a minimum, ET-1 and ANG II acting via the Rho/Rho-kinase and PPAR/NF-kappaB pathway. The induction of IL-18 in cardiomyocytes by Aldo, ET-1, and ANG II might, therefore, cause a deterioration of the cardiac function in an autocrine and paracrine fashion. The inhibition of the IL-18 expression by PPAR agonists might be one of the mechanisms whereby the beneficial cardiovascular effects are exerted.
Collapse
Affiliation(s)
- Takashi Doi
- Division of Coronary Heart Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya-City, Hyogo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Interleukin-18 enhances a Th2 biased response and susceptibility to Leishmania mexicana in BALB/c mice. Microbes Infect 2008; 10:834-9. [DOI: 10.1016/j.micinf.2008.03.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 03/20/2008] [Accepted: 03/21/2008] [Indexed: 11/23/2022]
|
27
|
Yoshimoto T, Yoshimoto T, Yasuda K, Mizuguchi J, Nakanishi K. IL-27 suppresses Th2 cell development and Th2 cytokines production from polarized Th2 cells: a novel therapeutic way for Th2-mediated allergic inflammation. THE JOURNAL OF IMMUNOLOGY 2007; 179:4415-23. [PMID: 17878337 DOI: 10.4049/jimmunol.179.7.4415] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
IL-27 up-regulates Th1 but down-regulates Th2 responses. However, its molecular mechanism and regulatory effects on polarized Th2 cells remain unclear. In this study, we have revealed that IL-27 inhibits Th2 cell development as well as Th2 cytokines production from already polarized Th2 cells by down-regulation of GATA-3 and up-regulation of T-bet expression simultaneously. In vivo daily IL-27 treatment for 1 wk after Leishmania major infection protects BALB/c mice from footpad swelling by diminishing parasite burden via reciprocal regulation of Th1 and Th2 responses. Furthermore, IL-27 stimulation causes marked reduction in the capacity of host mouse to mount a Th2 response against Strongyloides venezuelensis infection. Thus, IL-27-treated mice failed to develop intestinal mastocytosis after S. venezuelensis infection and exhibited a marked delay in parasite expulsion. Finally, intranasal administration of IL-27 inhibits OVA-induced airway hyperresponsiveness and inflammation in OVA-sensitized animals. Thus, IL-27 could provide us with a novel therapeutic way for treating Th2-associated diseases such as bronchial asthma.
Collapse
Affiliation(s)
- Tomohiro Yoshimoto
- Department of Immunology and Medical Zoology, Hyogo College of Medicine, Hyogo, Japan.
| | | | | | | | | |
Collapse
|
28
|
Arizono N, Nakanihsi K, Horii T, Tanabe K. Progress in the molecular biology of malaria and the immunology of nematode infections. Trends Parasitol 2007; 23:175-81. [PMID: 17320481 DOI: 10.1016/j.pt.2007.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 02/07/2007] [Accepted: 02/12/2007] [Indexed: 11/23/2022]
Abstract
Japan is one of a small number of countries to have successfully controlled or eliminated major parasitic diseases, including malaria, filariasis, schistosomiasis and enteric parasitoses. Of particular importance in this success was a close collaboration between primary research and public health efforts. Many Japanese researchers continue to study malaria, particularly the areas of genetics and immunology, and this should contribute to global parasite eradication strategies. Furthermore, studies of immunoregulation of nematode infection using the interleukin-18 pathway, most of which have been conducted in Japan, are helping to improve researchers' understanding of human immune mechanisms and host-parasite interactions.
Collapse
Affiliation(s)
- Naoki Arizono
- Department of Medical Zoology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kyoto 602-8566, Japan.
| | | | | | | |
Collapse
|
29
|
Carrada G, Cañeda C, Salaiza N, Delgado J, Ruiz A, Sanchez B, Gutiérrez-Kobeh L, Aguirre M, Becker I. Monocyte cytokine and costimulatory molecule expression in patients infected with Leishmania mexicana. Parasite Immunol 2007; 29:117-26. [PMID: 17266739 DOI: 10.1111/j.1365-3024.2006.00924.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Leishmania mexicana causes localized and diffuse cutaneous leishmaniasis. Patients with localized cutaneous leishmaniasis (LCL) develop a benign disease, whereas patients with diffuse cutaneous leishmaniasis (DCL) suffer from a progressive disease associated with anergy of the cellular response towards Leishmania antigens. We evaluated the production of the interleukins (IL) IL-12, IL-15, IL-18 and tumour necrosis factor-alpha (TNF-alpha) and the expression of the costimulatory molecules CD40, B7-1 and B7-2 in monocytes from LCL and DCL patients, stimulated in vitro with Leishmania mexicana lipophosphoglycan (LPG) for 18 h. LCL monocytes significantly increased TNF-alpha, IL-15 and IL-18 production, and this increase was associated with reduced amounts of IL-12. DCL monocytes produced no IL-15 or IL-18 and showed a decreasing tendency of TNF-alpha and IL-12 production as the severity of the disease increased. No difference was observed in the expression of CD40 and B7-1 between both groups of patients, yet B7-2 expression was significantly augmented in DCL patients. It remains to be established if this elevated B7-2 expression in DCL patients is cause or consequence of the Th2-type immune response that characterizes these patients. These data suggest that the diminished ability of the monocytes from DCL patients to produce cell-activating innate proinflammatory cytokines when stimulated with LPG is a possible cause for disease progression.
Collapse
Affiliation(s)
- G Carrada
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, México D.F., México
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
McDonald V, Pollok RCG, Dhaliwal W, Naik S, Farthing MJG, Bajaj-Elliott M. A potential role for interleukin-18 in inhibition of the development of Cryptosporidium parvum. Clin Exp Immunol 2006; 145:555-62. [PMID: 16907926 PMCID: PMC1809716 DOI: 10.1111/j.1365-2249.2006.03159.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence suggests that intestinal epithelial cells (IECs) constitutively express the immunoregulatory cytokine interleukin (IL)-18. IECs also serve as the host cell for the intracellular parasitic protozoan Cryptosporidium parvum. In the present study, C. parvum infection of a human enterocyte cell-line HCT-8 resulted in increased expression of IL-18 mRNA as measured by quantitative reverse transcription-polymerase chain reaction (RT-PCR). IL-18 protein was detected in control uninfected cells and following infection there was increased expression as measured by enzyme-linked immunosorbent assay (ELISA). Gene expression revealed the presence of the IL-18 receptor subunits not only in cell-lines but also in freshly isolated IECs, suggesting that IL-18-mediated signalling events may contribute to epithelial host defence during infection. Recombinant IL-18 inhibited intracellular development of the parasite in HCT-8 and HT-29 cells. Increased expression of bactericidal antibiotic peptides LL-37 and alpha-defensin 2 by IL-18 in HCT-8 and HT-29 cells may represent one mode of action by which this pluripotent cytokine aids in limiting the development of intracellular pathogens such as C. parvum in the gastrointestinal tract.
Collapse
Affiliation(s)
- V McDonald
- Barts and the London School of Medicine and Dentistry, Institute of Cell and Molecular Science, Centre for Gastroenterology, London, UK
| | | | | | | | | | | |
Collapse
|
31
|
Huang HY, Chan SH, Yu HT, Wang HS, Lai CH, Soong YK. Interleukin-18 system messenger RNA and protein expression in human endometrium during the menstrual cycle. Fertil Steril 2006; 86:905-13. [PMID: 17027359 DOI: 10.1016/j.fertnstert.2006.02.122] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2005] [Revised: 02/15/2006] [Accepted: 02/15/2006] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To investigate the IL-18 system, including IL-18, IL-18 receptor (IL-18R), IL-18 binding protein (IL-18BP), and IL-18 precursor (proIL-18), mRNA and protein expression in human endometrium during the menstrual cycle. DESIGN Controlled study. SETTING Clinical and academic research setting in a university medical center. PATIENT(S) Human endometrium was obtained from surgical specimens of normal cycling women undergoing hysterectomy for benign reasons. INTERVENTION(S) A total of 24 human endometrium samples were obtained at proliferative and secretory phase from surgical specimens of normal cycling women undergoing hysterectomy. Quantitative competitive polymerase chain reaction (QC PCR) and immunohistochemistry were performed. MAIN OUTCOME MEASURE(S) The differences of IL-18 system mRNA and the ratio of antagonist to agonist in both proliferative and secretory phases of endometrium were analyzed. RESULT(S) Our results showed a complete IL-18 system, including mRNA expression and protein production in both proliferative and secretory phases of the endometrium. The QC PCR demonstrated that both IL-18 and IL-18R decreased and IL-18BP increased mRNA expression in human endometrium in secretory phase compared with proliferative phase. A positive correlation of IL-18 and IL-18R in human endometrium was demonstrated in proliferative phase but not in secretory phase. In addition, there is a significantly higher ratio of IL-18BP to IL-18 mRNA levels in secretory endometrium compared with proliferative phase endometrium (P<.05). CONCLUSION(S) The expression and ratio of IL-18 antagonist to agonist may be responsible for embryo implantation.
Collapse
Affiliation(s)
- Hong-Yuan Huang
- Department of Obstetrics and Gynecology, Lin-Kou Medical Center, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.
| | | | | | | | | | | |
Collapse
|
32
|
Rowland CA, Lertmemongkolchai G, Bancroft A, Haque A, Lever MS, Griffin KF, Jackson MC, Nelson M, O'Garra A, Grencis R, Bancroft GJ, Lukaszewski RA. Critical role of type 1 cytokines in controlling initial infection with Burkholderia mallei. Infect Immun 2006; 74:5333-40. [PMID: 16926428 PMCID: PMC1594859 DOI: 10.1128/iai.02046-05] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Revised: 02/07/2006] [Accepted: 04/14/2006] [Indexed: 01/14/2023] Open
Abstract
Burkholderia mallei is a gram-negative bacterium which causes the potentially fatal disease glanders in humans; however, there is little information concerning cell-mediated immunity to this pathogen. The role of gamma interferon (IFN-gamma) during B. mallei infection was investigated using a disease model in which infected BALB/c mice normally die between 40 and 60 days postinfection. IFN-gamma knockout mice infected with B. mallei died within 2 to 3 days after infection, and there was uncontrolled bacterial replication in several organs, demonstrating the essential role of IFN-gamma in the innate immune response to this pathogen. Increased levels of IFN-gamma, interleukin-6 (IL-6), and monocyte chemoattractant protein 1 were detected in the sera of immunocompetent mice in response to infection, and splenic mRNA expression of IFN-gamma, IL-6, IL-12p35, and IL-27 was elevated 24 h postinfection. The effects of IL-18, IL-27, and IL-12 on stimulation of the rapid IFN-gamma production were investigated in vitro by analyzing IFN-gamma production in the presence of heat-killed B. mallei. IL-12 was essential for IFN-gamma production in vitro; IL-18 was also involved in induction of IFN-gamma, but IL-27 was not required for IFN-gamma production in response to heat-killed B. mallei. The main cellular sources of IFN-gamma were identified in vitro as NK cells, CD8+ T cells, and TCRgammadelta T cells. Our data show that B. mallei is susceptible to cell-mediated immune responses which promote expression of type 1 cytokines. This suggests that development of effective vaccines against glanders should target the production of IFN-gamma.
Collapse
Affiliation(s)
- Caroline A Rowland
- Biomedical Sciences, Dstl, Porton Down, Salisbury, SP4 0JQ, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Basophils and mast cells are effecter cells in allergen/IgE-mediated immune responses. They induce type 1 immediate immune response in airway or other organ, resulting in bronchial asthma and other allergic diseases. However, they also play a critical role in host defense against infection with helminthes. Upon linkage of FcepsilonRI with a complex of allergen and IgE, basophils and mast cells release a large amount of Th2 cytokines and chemical mediators. Therefore these responses are "acquired allergic responses" and induce allergic diseases, such as bronchial asthma. However, basophils and mast cells derived from cultured bone marrow cells with IL-3 for 10 days express IL-18Ralpha chain and produce Th2 cytokines in response to the stimulation with IL-3 and IL-18 without FcepsilonRI cross-linkage. Furthermore, they produce Th2 cytokines upon stimulation with several TLR ligands, such as LPS. This finding may suggest the presence of allergen/IgE-independent allergic responses, which we would like to designate as "innate allergic response". However, in vivo treatment with IL-18 and IL-2 protects against gastrointestinal nematode infection by activating intestinal mucosal mast cells in STAT6-independent manner, suggesting the importance of innate allergic response against helminth infection. Here we discuss the functional role of IL-18-induced "innate allergic response" in disease and host defense.
Collapse
Affiliation(s)
- Tomohiro Yoshimoto
- Department of Immunology and Medical Zoology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | | |
Collapse
|
34
|
Zaitsu M, Yamasaki F, Ishii E, Midoro-Horiuti T, Goldblum RM, Hamasaki Y. Interleukin-18 primes human basophilic KU812 cells for higher leukotriene synthesis. Prostaglandins Leukot Essent Fatty Acids 2006; 74:61-6. [PMID: 16280246 DOI: 10.1016/j.plefa.2005.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Revised: 09/13/2005] [Accepted: 09/26/2005] [Indexed: 10/25/2022]
Abstract
The human basophilic cell line KU812 that is an established tool for studying the function of human basophils, is differentiated into mature basophils by interleukin (IL-3) or other agents. However, whether leukotrienes (LTs)-synthesis is affected by cytokines in KU812 cells remains unknown. KU812 cells were incubated with IL-3, IL-4, IL-6, IL-13 or IL-18 for up to 14 days. The A23187 stimulated- and IgE cross-linked-synthesis of LTC(4) and LTB(4) were measured using an enzyme immunoassay (EIA). The expression of messenger RNA (mRNA) for LT-synthesizing enzymes was examined by reverse transcriptase polymerase chain reaction (RT-PCR), and the expression of 5-lipoxygenase (5-LO) was examined by immunostaining. Incubation with IL-3 (10 ng/ml) and IL-18 (10 ng/ml) induced the expression of 5-LO. A23187stimulated LT-synthesis and IgE cross-linked LT-synthesis were enhanced after incubation with IL-3 or IL-18. These results indicated that IL-3 and IL-18 primed human basophils for higher LT-synthesis. Thus, both IL-3 and IL-18 might be important factors for regulating LT-synthesis during the differentiation of human basophils.
Collapse
Affiliation(s)
- Masafumi Zaitsu
- Department of Pediatrics, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga City 849-8501, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Sasaki Y, Yoshimoto T, Maruyama H, Tegoshi T, Ohta N, Arizono N, Nakanishi K. IL-18 with IL-2 protects against Strongyloides venezuelensis infection by activating mucosal mast cell-dependent type 2 innate immunity. ACTA ACUST UNITED AC 2005; 202:607-16. [PMID: 16129701 PMCID: PMC2212862 DOI: 10.1084/jem.20042202] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
C57BL/6 (B6) and B6 background STAT6−/− mice pretreated with IL-18 plus IL-2 showed prominent intestinal mastocytosis and rapidly expelled implanted adult worms of the gastrointestinal nematode Strongyloides venezuelensis. In contrast, identically pretreated mast cell–deficient W/Wv mice failed to do so. Thus, activated mucosal mast cells (MMC) are crucial for parasite expulsion. B6 mice infected with S. venezuelensis third-stage larvae (L3) completed parasite expulsion by day 12 after infection, whereas IL-18−/− or IL-18Rα−/− B6 mice exhibited marked impairment in parasite expulsion, suggesting a substantial contribution of IL-18–dependent MMC activation to parasite expulsion. Compared with IL-18−/− or IL-18Rα−/− mice, S. venezuelensis L3–infected STAT6−/− mice have poorly activated MMC and sustained infection; although their IL-18 production is normal. Neutralization of IL-18 and IL-2 further reduces expulsion in infected STAT6−/− mice. These results suggest that collaboration between IL-18–dependent and Th2 cell–dependent mastocytosis is important for prompt parasite expulsion.
Collapse
Affiliation(s)
- Yuki Sasaki
- Department of Immunology and Medical Zoology, Hyogo College of Medicine, Nishinomiya, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Matsui K, Tsutsui H, Nakanishi K. Pathophysiological roles for IL-18 in inflammatory arthritis. Expert Opin Ther Targets 2005; 7:701-24. [PMID: 14640907 DOI: 10.1517/14728222.7.6.701] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
IL-18 is a unique cytokine with prominently wide spectrum biological actions. Among these, its IFN-gamma/TNF-alpha-inducing activity primarily contributes to the development of various inflammatory diseases including inflammatory arthritis. IL-18 levels correlate with the disease activity of rheumatoid arthritis (RA) and osteoarthritis (OA). IL-18 is spontaneously released from RA synovial cells and OA chondrocytes and seems to participate in the development of the inflammatory and destructive alterations of joints via induction of TNF-alpha, a potent effector molecule. TNF-alpha, in turn, increases IL-18 expression in RA synovial cells. Recent clinical trials have revealed the efficacy of TNF-alpha in RA with a reduction in circulatory IL-18 levels. These may implicate the positive circuit between IL-18 and TNF-alpha for development of RA. As IL-18-deficient mice evade collagen-induced arthritis in a mouse RA model, therapeutics targeting IL-18 may be beneficial against RA/OA. Here, the authors review the possible roles of IL-18 in inflammatory arthritis.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibody Formation
- Arthritis, Experimental/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/physiopathology
- Arthritis, Rheumatoid/therapy
- Autoimmune Diseases/immunology
- Autoimmune Diseases/physiopathology
- Autoimmune Diseases/therapy
- Chondrocytes/metabolism
- Cysteine Endopeptidases/physiology
- Cytokines/physiology
- Humans
- Immunity, Cellular
- Infections/immunology
- Inflammation/physiopathology
- Interleukin-18/antagonists & inhibitors
- Interleukin-18/deficiency
- Interleukin-18/genetics
- Interleukin-18/physiology
- Interleukin-18 Receptor alpha Subunit
- Membrane Glycoproteins/drug effects
- Membrane Glycoproteins/physiology
- Mice
- Mice, Knockout
- Models, Immunological
- Neoplasms/immunology
- Osteoarthritis/immunology
- Osteoarthritis/physiopathology
- Osteoarthritis/therapy
- Receptors, Cell Surface/drug effects
- Receptors, Cell Surface/physiology
- Receptors, Interleukin/drug effects
- Receptors, Interleukin/physiology
- Receptors, Interleukin-18
- Signal Transduction/physiology
- Synovial Membrane/metabolism
- Th1 Cells/immunology
- Th2 Cells/immunology
- Toll-Like Receptors
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- Kiyoshi Matsui
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | | | | |
Collapse
|
37
|
Fernández-Lago L, Orduña A, Vizcaíno N. Reduced interleukin-18 secretion in Brucella abortus 2308-infected murine peritoneal macrophages and in spleen cells obtained from B. abortus 2308-infected mice. J Med Microbiol 2005; 54:527-531. [PMID: 15888459 DOI: 10.1099/jmm.0.45936-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Th1 immune responses in which gamma interferon (IFN-gamma) production predominates are associated with protective immunity against intracellular bacteria. Following infection, interleukin-18 (IL-18) may contribute, in association with IL-12, to optimal IFN-gamma production. In this study, the secretion of IL-18 following intracellular infection with virulent Brucella abortus 2308 in CD-1 cultured peritoneal macrophages and splenocyte cultures was investigated. The production of IL-18 was reduced in both CD-1 mouse peritoneal macrophages infected with B. abortus 2308 and splenocyte cultures obtained from B. abortus 2308-infected mice at 3, 6 and 10 days post-infection (p.i.). In contrast, splenocyte cultures obtained from B. abortus 2308-infected mice at 3 days p.i. secreted significant amounts of IFN-gamma. Stimulation of these cells with recombinant IL-18 (rIL-18) and/or rIL-12 did not significantly increase IFN-gamma secretion at the splenocyte level. These data suggest that once the infection has been established, B. abortus 2308 selectively limits IL-18 secretion without affecting endogenous IFN-gamma production.
Collapse
Affiliation(s)
- Luis Fernández-Lago
- Departamento de Microbiología y Genética, Edificio Departamental, Universidad de Salamanca, Plaza Doctores de la Reina s/n, 37007 Salamanca, Spain 2Departamento de Microbiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| | - Antonio Orduña
- Departamento de Microbiología y Genética, Edificio Departamental, Universidad de Salamanca, Plaza Doctores de la Reina s/n, 37007 Salamanca, Spain 2Departamento de Microbiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| | - Nieves Vizcaíno
- Departamento de Microbiología y Genética, Edificio Departamental, Universidad de Salamanca, Plaza Doctores de la Reina s/n, 37007 Salamanca, Spain 2Departamento de Microbiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
38
|
Day AS, Su B, Ceponis PJM, Jones NL, Yau E, Sieveking D, Sherman PM. Helicobacter pylori infection induces interleukin-18 production in gastric epithelial (AGS) cells. Dig Dis Sci 2004; 49:1830-1835. [PMID: 15628712 DOI: 10.1007/s10620-004-9579-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Helicobacter pylori colonization of the stomach results in a chronic-active gastritis characterized by mucosal infiltration of both neutrophils and lymphocytes. A T helper lymphocyte (Th1) profile predominates, which promotes the chronic and persistent inflammatory changes in the gastric mucosa in response to this bacterial pathogen. The cytokine interleukin-18 induces production of interferon-gamma by activated T lymphocytes and promotes a Th1 profile. An in vitro model system was utilized to determine the role of interleukin-18 in response to infection of gastric epithelial cells by H. pylori. H. pylori isolates, characterized with respect to cagE and cagA and VacA status, were employed to infect AGS gastric epithelial cells. Interleukin-18 production was determined by immunoassay. Infection of AGS cells with H. pylori resulted in a 1.8-fold increase in interleukin-18 compared to uninfected cells (22.7+/-2.4 vs. 12.7+/-2.2 pg/ml; P < 0.005). This interleukin-18 response was independent of the cagE status of infecting strains (23.3+/-1.9 vs. 26.3+/-3.6 pg/ml; P = NS). Exposure of AGS cells to recombinant interleukin-18 resulted in dose-dependent and time-dependent secretion of interleukin-8 that was maximal following exposure to 100 pg/ml interleukin-18 for 24 hr (292+/-5 pg/ml, versus 102+/-14 pg/ml in unstimulated cells; P < 0.001). Interleukin-8 secretion was inhibited following pretreatment of cells with anti-interleukin-18 antibody and by pharmacological inhibition of the nuclear transcription factor, NF-kappaB. These findings demonstrate that interleukin-18 can enhance host chemokine response to H. pylori infection.
Collapse
Affiliation(s)
- Andrew S Day
- Sydney Children's Hospital and School of Women's and Children's Health, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | |
Collapse
|
39
|
Gomes-Pereira S, Rodrigues OR, Rolão N, Almeida PD, Santos-Gomes GM. Hepatic cellular immune responses in mice with "cure" and "non-cure" phenotype to Leishmania infantum infection: importance of CD8+ T cells and TGF-beta production. ACTA ACUST UNITED AC 2004; 41:59-68. [PMID: 15094168 DOI: 10.1016/j.femsim.2004.01.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2003] [Revised: 12/15/2003] [Accepted: 01/12/2004] [Indexed: 11/30/2022]
Abstract
The objective of this study was to analyse hepatic cellular immune response of mice with "cure" and "non-cure" phenotypes to Leishmania infantum infection. During infection establishment, elevated TGF-beta levels and absence of a Th1 response may have contributed to parasite multiplication and to similar hepatic parasitic loads. Later in infection, an increase in the number and activation levels of CD8+ cells was observed simultaneously with parasite elimination, but only significant in "cure" strain. During this recovering phase, "non-cure" animals showed low Th2 cytokine levels, while TGF-beta production was higher than in "cure" mice. These results point out to a role for CD8+ T cells in liver acquired immune response and to TGF-beta regulation of "cure" and "non-cure" phenotype to L. infantum infection.
Collapse
Affiliation(s)
- Sandra Gomes-Pereira
- Unidade de Leishmanioses, Centro de Malária e Outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Rua da Junqueira 96, 1349-008 Lisboa, Portugal
| | | | | | | | | |
Collapse
|
40
|
McMahon-Pratt D, Alexander J. Does the Leishmania major paradigm of pathogenesis and protection hold for New World cutaneous leishmaniases or the visceral disease? Immunol Rev 2004; 201:206-24. [PMID: 15361243 DOI: 10.1111/j.0105-2896.2004.00190.x] [Citation(s) in RCA: 235] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Parasitic protozoa of the genus Leishmania have provided a useful perspective for immunologists in terms of host defense mechanisms critical for the resolution of infection caused by intracellular pathogens. These organisms, which normally reside in a late endosomal, major histocompatibility complex (MHC) class II(+) compartment within host macrophages cells, require CD4(+) T-cell responses for the control of disease. The paradigm for the CD4(+) T-helper 1 (Th1)/Th2 dichotomy is largely based on the curing/non-curing responses, respectively, to Leishmania major infection. However, this genus of parasitic protozoa is evolutionarily diverse, with the cutaneous disease-causing organisms of the Old World (L. major) and New World (Leishmania mexicana/ Leishmania amazonensis) having diverged 40-80 million years ago. Further adaptations to survive within the visceral organs (for Leishmania donovani, Leishmania chagasi, and Leishmania infantum) must have been required. Consequently, significant differences in host-parasite interactions have evolved. Different virulence factors have been identified for distinct Leishmania species, and there are profound differences in the immune mechanisms that mediate susceptibility/resistance to infection and in the pathology associated with disease. These variations not only point to interesting features of the host-pathogen interaction and immunobiology of this genus of parasitic protozoa, but also have important implications for immunotherapy and vaccine development.
Collapse
Affiliation(s)
- Diane McMahon-Pratt
- Department of Epidemiology & Public Health, Yale University School of Medicine, New Haven, CT, USA.
| | | |
Collapse
|
41
|
Akhiani AA, Schön K, Lycke N. Vaccine-induced immunity against Helicobacter pylori infection is impaired in IL-18-deficient mice. THE JOURNAL OF IMMUNOLOGY 2004; 173:3348-56. [PMID: 15322198 DOI: 10.4049/jimmunol.173.5.3348] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Protective immunity against Helicobacter pylori infection in mice has been associated with a strong Th1 response, involving IL-12 as well as IFN-gamma, but recent studies have also demonstrated prominent eosinophilic infiltration, possibly linked to local Th2 activity in the gastric mucosa. In this study we investigated the role of IL-18, because this cytokine has been found to be a coregulator of Th1 development as well as involved in Th2-type responses with local eotaxin production that could influence gastric eosinophilia and resistance to infection. We found that IL-18(-/-) mice failed to develop protection after oral immunization with H. pylori lysate and cholera toxin adjuvant, indicating an important role of IL-18 in protection. Well-protected C57BL/6 wild-type (WT) mice demonstrated substantial influx of CD4(+) T cells and eosinophilic cells in the gastric mucosa, whereas IL-18(-/-) mice had less gastritis, few CD4(+) T cells, and significantly reduced numbers of eosinophilic cells. T cells in well-protected WT mice produced increased levels of IFN-gamma and IL-18 to recall Ag. By contrast, unprotected IL-18(-/-) mice exhibited significantly reduced gastric IFN-gamma and specific IgG2a Ab levels. Despite differences in gastric eosinophilic cell infiltration, protected WT and unprotected IL-18(-/-) mice had comparable levels of local eotaxin, suggesting that IL-18 influences protection via Th1 development and IFN-gamma production rather than through promoting local production of eotaxin and eosinophilic cell infiltration.
Collapse
Affiliation(s)
- Ali A Akhiani
- Department of Clinical Immunology, Goteborg University, Goteborg, Sweden.
| | | | | |
Collapse
|
42
|
Li Y, Ishii K, Hisaeda H, Hamano S, Zhang M, Nakanishi K, Yoshimoto T, Hemmi H, Takeda K, Akira S, Iwakura Y, Himeno K. IL-18 gene therapy develops Th1-type immune responses in Leishmania major-infected BALB/c mice: is the effect mediated by the CpG signaling TLR9? Gene Ther 2004; 11:941-8. [PMID: 14985787 DOI: 10.1038/sj.gt.3302240] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
IL-18 regulates either Th1 or Th2 responses depending on the cytokine microenvironment. Administration of recombinant IL-18 (rIL-18) alone does not promote Th1 response, but rather induces Th2 response and exacerbates Leishmania major infection in susceptible BALB/c mice. Here, we treated BALB/c mice with an IL-18-expressing plasmid by using a gene gun weekly after L. major infection. This gene therapy resulted in improved pathogenic process and preferential induction of Th1 responses by inducing the expression of IL-12 p40, but treatment with rIL-18 did not. Notably, simultaneous administration of rIL-18 with an empty plasmid vector rendered BALB/c mice resistant to the infection, despite the fact that treatment with either rIL-18 alone or the plasmid vector alone did not influence the susceptibility. The synergistic role of the vector with rIL-18 was found to depend on CpG motifs, which enhanced expression of proinflammatory cytokines, especially IL-12, from APCs through Toll-like receptor (TLR) 9 ligation. Treatment with methylated plasmid vector in which CpG was disrupted could no longer prevent the disease development in coadministration with rIL-18. Taken together, IL-18 gene therapy was shown to develop Th1-type protective immunity in L. major-infected BALB/c mice without the requirement of exogenous IL-12, probably via CpG-TLR9 signaling pathway.
Collapse
Affiliation(s)
- Y Li
- Department of Microbiology and Immunology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wei XQ, Niedbala W, Xu D, Luo ZX, Pollock KGJ, Brewer JM. Host genetic background determines whether IL-18 deficiency results in increased susceptibility or resistance to murine Leishmania major infection. Immunol Lett 2004; 94:35-7. [PMID: 15234532 DOI: 10.1016/j.imlet.2004.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Revised: 04/08/2004] [Accepted: 04/08/2004] [Indexed: 10/26/2022]
Abstract
Interleukin-18 (IL-18) plays an important role in innate and acquired immunity. IL-18 gene deficient (IL-18-/-) mice of the 129 x CD1 strain were reported to be more susceptible to Leishmania major infection than the wild-type mice. In contrast IL-18-/- mice of the C57BL/6 background were found to be as resistant as the wild-type (WT) mice. To resolve this discrepancy, IL-18 gene deficiency was introduced by backcrossing on to the highly susceptible BALB/c, or the moderately resistant DBA/1 backgrounds. Here we have demonstrated that BALB/c IL-18-/- mice were more resistant to L. major infection than WT BALB/c mice, whereas DBA/1 IL-18-/- mice were markedly more susceptible than their WT littermates. BALB/c IL-18-/- mice produced less IFNgamma and IL-4, whereas DBA/1 IL-18ko mice produced more IFNgamma and IL-4 than their respective WT controls. These result clearly demonstrate that the role of IL-18 in resistance or susceptibility to L. major is determined by host genetic background.
Collapse
Affiliation(s)
- Xiao-Qing Wei
- Division of Immunology, Infection and Inflammation, University of Glasgow, Western Infirmary, Glasgow, Scotland G11 6NT, UK.
| | | | | | | | | | | |
Collapse
|
44
|
Chung SW, Choi SH, Kim TS. Induction of persistent in vivo resistance to Mycobacterium avium infection in BALB/c mice injected with interleukin-18-secreting fibroblasts. Vaccine 2004; 22:398-406. [PMID: 14670321 DOI: 10.1016/j.vaccine.2003.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Interferon-gamma (IFN-gamma) is closely associated with the generation of cell-mediated immunity and resistance to intracellular parasites. Interleukin-18 (IL-18) is known to strongly induce IFN-gamma production by T cells and natural killer (NK) cells. To determine whether the paracrine secretion of IL-18 can efficiently stimulate the resistance to Mycobacterium avium complex (MAC) infection, 3T3 fibroblasts were stably transfected to secrete bioactive IL-18 and their effects on MAC infection were investigated in genetically susceptible BALB/c mice, compared with that of free recombinant IL-18. Immunization with IL-18-secreting fibroblasts (3T3/IL-18) during intranasal infection with MAC resulted in a significant decrease in bacterial load of lung during the entire 8-week observation period, while rIL-18 reduced the bacterial load at initial 1 week but not by 8 weeks postinfection. Immunization with the 3T3/IL-18 cells induced and maintained significantly higher levels of cytotoxic activity and nitric oxide production by lung cells than those of rIL-18 immunization. Furthermore, lung cells in mice injected with the 3T3/IL-18 cells showed persistent production of IFN-gamma throughout the 8-week period, suggesting that the 3T3/IL-18 cells induced the resistance to MAC infection via IFN-gamma production. This work suggests that IL-18-secreting fibroblasts may serve as a vehicle for paracrine secretion of IL-18 in immunotherapy of MAC infection.
Collapse
Affiliation(s)
- Su W Chung
- Department of Pharmacy, College of Pharmacy, Chonnam National University, 500-757 Kwangju, Republic of Korea
| | | | | |
Collapse
|
45
|
Netea MG, Kullberg BJ, Van der Meer JWM. Proinflammatory Cytokines in the Treatment of Bacterial and Fungal Infections. BioDrugs 2004; 18:9-22. [PMID: 14733604 DOI: 10.2165/00063030-200418010-00002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mortality due to severe bacterial infections has not been markedly effected by the introduction of new antimicrobial drugs over the last 30-40 years. This has emphasized the need for development of new therapeutic strategies to combat sepsis. The outcome of an infection depends on two factors: the growth of the microorganisms (including the effect of antibacterial drugs), and the host's defensive response to the invading organism. It is known that injection of bacterial products into experimental animals leads to enhanced nonspecific resistance to a variety of microorganisms. The discovery of the specific mediators responsible for modulation of host defense has created new possibilities for the development of alternative treatment strategies. Molecules such as interleukins, interferons, tumor necrosis factors and hematopoietic growth factors have become available in recombinant form, and their therapeutic potential in various infectious diseases has been tested in various experimental models of infections. Initial data in various patient groups indicate that adjunctive therapy with recombinant proinflammatory cytokines may have beneficial effects in the treatment of bacterial and fungal infections.
Collapse
Affiliation(s)
- Mihai G Netea
- Department of Medicine, University Medical Center St. Radboud, Nijmegen, The Netherlands
| | | | | |
Collapse
|
46
|
Kawakami K. Promising immunotherapies with Th1-related cytokines against infectious diseases. J Infect Chemother 2003; 9:201-9. [PMID: 14513386 DOI: 10.1007/s10156-003-0263-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2003] [Indexed: 11/29/2022]
Abstract
In recent years, there has been an increase in the number of individuals with compromised immune systems. This is due to the rise in the numbers of aging people, patients receiving immunosuppressive treatment after organ transplantation, patients with hematological malignancies, and patients with AIDS. These individuals frequently fall into helper T cell (Th)1-Th2 cytokine imbalance due to a shift towards a Th2-dominant condition. Such a pathological condition puts them at a high risk for developing infectious diseases caused by a variety of microbial pathogens which are often refractory to conventional chemotherapy. Therefore, the administration of Th1-related cytokines is expected to be promising immunotherapy against these intractable infectious diseases. In a series of investigations, we have demonstrated the effectiveness of treatment with Th1-related cytokines, such as interferon (IFN)-gamma, interleukin (IL)-12, and IL-18, in protecting animals from experimental infectious diseases caused by Mycobacterium tuberculosis and Cryptococcus neoformans. Recently, several investigators reported successful clinical treatment with IFN-gamma or IL-12 in patients with intractable tuberculous and nontuberculous mycobacteriosis. Thus, now is an appropriate time for scientific evaluation to clinically confirm the effectiveness of these novel immunotherapies.
Collapse
Affiliation(s)
- Kazuyoshi Kawakami
- Division of Infectious Diseases, Department of Internal Medicine, Graduate School and Faculty of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan.
| |
Collapse
|
47
|
Pfaff AW, Kirch AK, Hoffmann WH, Banla M, Schulz-Key H, Geiger SM, Soboslay PT. Regulatory effects of IL-12 and IL-18 on Onchocerca volvulus- and Entamoeba histolytica-specific cellular reactivity and cytokine profiles. Parasite Immunol 2003; 25:325-32. [PMID: 14507330 DOI: 10.1046/j.1365-3024.2003.00638.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the present study, the cytokines interleukin (IL)-12 and IL-18 were evaluated for their capacity to modulate and to re-direct in vitro parasite antigen-specific cellular responsiveness in patients exposed to Onchocerca volvulus and Entamoeba histolytica infection. We found that IL-18 was highly capable of reducing parasite antigen-induced IL-10 production by PBMC. In contrast, addition or neutralization of IL-12, also in combination with IL-18 and the interferon-gamma-inducible chemokine IP-10 did not affect IL-10 production. Interestingly, the highest IL-10 levels were measured when IL-18 and IP-10 were both neutralized. Although having no effect on IL-10, IL-12 strongly promoted spontaneous and parasite antigen-driven IFN-gamma production by PBMC, whereas IL-18 was only moderately affecting IFN-gamma release by PBMC re-stimulated with E. histolytica- or O. volvulus-specific antigens. Both IL-12 and IL-18 diminished the cellular production of IL-13, and a synergistic effect was observed when the cytokines were combined. Likewise, neutralization of IL-12 enhanced Entamoeba and Onchocerca antigen-driven IL-13 production, but no further increase of IL-13 was observed, when anti-IL-12 and anti-IL-18 were used together. This study disclosed that IL-18 will significantly down-regulate parasite-specific IL-10 production, whereas IL-12 induced IFN-gamma and inhibited IL-13 production by PBMC from humans exposed to O. volvulus and E. histolytica. Such selective immune-regulatory capacity of IL-12 and IL-18 may comprise an important tool to re-direct polarized cytokine responses towards a balanced Th1/Th2 cytokine profile, which may prevent pathology and promote immunity against helminth and protozoan parasite infections.
Collapse
|
48
|
Dumas C, Muyombwe A, Roy G, Matte C, Ouellette M, Olivier M, Papadopoulou B. Recombinant Leishmania major secreting biologically active granulocyte-macrophage colony-stimulating factor survives poorly in macrophages in vitro and delays disease development in mice. Infect Immun 2003; 71:6499-509. [PMID: 14573672 PMCID: PMC219543 DOI: 10.1128/iai.71.11.6499-6509.2003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leishmania is an intracellular pathogen that replicates inside macrophages. Activated macrophages produce a specific subset of cytokines that play an important role in the control of Leishmania infections. As part of our interest in developing suicide parasites that produce abortive infections for the purposes of vaccination, we engineered recombinant Leishmania major strains producing biologically active granulocyte-macrophage colony-stimulating factor (GM-CSF). We showed that GM-CSF is being produced in the phagosomes of infected macrophages and that it can be detected in the culture supernatants of both infected macrophages and extracellular parasites. Our data support the notion that GM-CSF secreted by both developmental forms of recombinant L. major can activate macrophages to produce high levels of proinflammatory cytokines such as interleukin-1beta (IL-1beta), IL-6, and IL-18 and various chemokines including RANTES/CCL5, MIP-1alpha/CCL3, MIP-1beta/CCL4, MIP-2/CXCL2, and MCP-1/CCL2, which enhance parasite killing. Indeed, GM-CSF-expressing parasites survive poorly in macrophages in vitro and produce delayed lesion development in susceptible BALB/c mice in vivo. Selective killing of intracellular Leishmania expressing cytokine genes capable of activating cellular responses may constitute a promising strategy to control and/or prevent parasitic infections.
Collapse
Affiliation(s)
- Carole Dumas
- Department of Medical Biology, Faculty of Medicine, Laval University, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Remick DG, Bolgos GE, Siddiqui J. Inflammatory status in sepsis alters efficacy of interleukin-18 binding protein therapy. Crit Care Med 2003; 31:2096-101. [PMID: 12973165 DOI: 10.1097/01.ccm.0000080492.81509.29] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Sepsis remains a serious clinical problem, and multiple attempts at blocking inflammation have failed to decrease mortality rate. Interleukin-18 has been demonstrated to be an important component of the innate immune response to bacterial infections. DESIGN Previous work demonstrated that elevated plasma concentrations of interleukin-6 obtained in the first 6 hrs of sepsis predict a worse outcome. Mice were subjected to cecal ligation and puncture and, on the basis of the plasma concentration of interleukin-6, were randomized to receive either interleukin-18 binding protein or vehicle approximately 8 hrs after the onset of sepsis. SETTING University research laboratory. SUBJECTS Adult, female BALB/c mice. INTERVENTIONS We sought to determine the role of interleukin-18 in sepsis by blocking its biological activity with the interleukin-18 binding protein in the murine model of sepsis induced by cecal ligation and puncture. MEASUREMENTS AND MAIN RESULTS In this study, elevated plasma concentrations of interleukin-6 were associated with a worse outcome. Treatment with interleukin-18 binding protein decreased inflammation as determined by lower concentrations of plasma interleukin-6 obtained 48 hrs after the onset of sepsis. In mice with increased risk of dying, interleukin-18 binding protein slightly decreased mortality rate. However, in those mice with a predicted low mortality rate, interleukin-18 binding protein significantly increased mortality rate. CONCLUSIONS In this study, mice at low risk of death due to sepsis had decreased survival when treated with interleukin-18 binding protein. These results have potential implications for the use of interleukin-18 binding protein for treatment of chronic inflammatory conditions since it may place the host at increased risk of infectious complications.
Collapse
Affiliation(s)
- Daniel G Remick
- Department of Pathology, University of Michigan, Ann Arbor, 48109-0602, USA.
| | | | | |
Collapse
|
50
|
Graefe SEB, Jacobs T, Gaworski I, Klauenberg U, Steeg C, Fleischer B. Interleukin-12 but not interleukin-18 is required for immunity to Trypanosoma cruzi in mice. Microbes Infect 2003; 5:833-9. [PMID: 12919851 DOI: 10.1016/s1286-4579(03)00176-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Protective immunity to the parasite Trypanosoma cruzi in mice depends on a pro-inflammatory T cell response involving the production of interferon-gamma (IFN-gamma). In conjunction with interleukin-12 (IL-12), IL-18 promotes the synthesis of IFN-gamma and a T helper type 1 immune response. We investigated the requirements of IL-12 and IL-18 in murine T. cruzi infection by use of C57BL/6 mice genetically deficient in either cytokine. IL-12p40(-/-) mice succumbed to infection at doses of 100 parasites, whereas IL-18(-/-) and wild-type mice resisted infectious doses up to 1000 parasites to the same extent. Levels of parasitemia were comparable between the latter groups, as were tissue parasite burdens according to quantitative real-time PCR. In contrast, IL-12p40(-/-) mice displayed vastly increased levels of parasites both in blood and in tissue. IFN-gamma concentrations in the serum of infected mice and in supernatants of splenocytes stimulated in vitro were decreased in IL-18(-/-) mice, whereas in IL-12p40(-/-) mice, IFN-gamma was undetectable in the serum and drastically reduced in cell supernatants. Levels of IL-12 production were generally comparable between wild-type and IL-18(-/-) mice, as were levels of IL-4, IL-2 and nitric oxide. Thus, the requirement for endogenous pro-inflammatory cytokines for a protective murine immune response against T. cruzi is satisfied by the expression of IL-12, while IL-18 is dispensable.
Collapse
Affiliation(s)
- Sebastian E B Graefe
- Department for Medical Microbiology and Immunology, Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany.
| | | | | | | | | | | |
Collapse
|