1
|
Hendrix EK, Sha J, Kilgore PB, Neil BH, Chopra AK. Combination of live attenuated and adenovirus-based vaccines completely protects interferon gamma (IFNγ) knockout mice against pneumonic plague. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627261. [PMID: 39713400 PMCID: PMC11661069 DOI: 10.1101/2024.12.06.627261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Two live attenuated vaccines (LAVs), LMA and LMP, were evaluated alone or in combination with a trivalent adenoviral vector-based vaccine (Ad5-YFV) for their efficacy and immune responses in wild type (WT) and interferon gamma (IFNγ) knockout (KO) mice in a C57BL/6 background. While LMA and LMP are triple deletion mutants of Yersinia pestis CO92 strain, Ad5-YFV incorporates three protective plague immunogens. An impressive 80-100% protection was observed in all vaccinated animals against highly lethal intranasal challenge doses of parental Y. pestis CO92. All vaccinated mice generated robust humoral and cellular immune responses. The immunized WT mice showed overall better antibody responses in both serum and bronchoalveolar lavage fluid with much higher percentages of polyfunctional T cell populations. On the other hand, vaccinated IFNγ KO mice displayed better B cell activity in germinal centers with higher percentages of activated antigen specific T cells and memory T cells. In addition, depletion of IFNγ and tumor necrosis factor alpha (TNFα) from immunized WT mice prior to and during infection did not reduce protection against pulmonary Y. pestis CO92 challenge. These data demonstrated a dispensable nature of IFNγ in mediating protection by the aforementioned vaccines. This is the first detailed immunogenicity study of two plague LAVs administered either alone or in combination with an Ad5-YFV vaccine in a prime-boost immunization strategy in IFNγ KO mice. Further, by combining advantages of live-attenuated and adenovirus-based vaccines, augmentation of generalized immune responses were observed which could be beneficial in providing long-lasting immunity in the host.
Collapse
|
2
|
Casanova JL, MacMicking JD, Nathan CF. Interferon- γ and infectious diseases: Lessons and prospects. Science 2024; 384:eadl2016. [PMID: 38635718 DOI: 10.1126/science.adl2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/13/2024] [Indexed: 04/20/2024]
Abstract
Infectious diseases continue to claim many lives. Prevention of morbidity and mortality from these diseases would benefit not just from new medicines and vaccines but also from a better understanding of what constitutes protective immunity. Among the major immune signals that mobilize host defense against infection is interferon-γ (IFN-γ), a protein secreted by lymphocytes. Forty years ago, IFN-γ was identified as a macrophage-activating factor, and, in recent years, there has been a resurgent interest in IFN-γ biology and its role in human defense. Here we assess the current understanding of IFN-γ, revisit its designation as an "interferon," and weigh its prospects as a therapeutic against globally pervasive microbial pathogens.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, 75015 Paris, France
| | - John D MacMicking
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, Yale University, West Haven, CT 06477, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Carl F Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
3
|
Hwang YH, Byeon Y, Ahn SH, Kim MY, Byun SH, Lee HJ, Suh B, Kim D, Jung EJ, Kim YJ. Live attenuated smallpox vaccine candidate (KVAC103) efficiently induces protective immune responses in mice. Vaccine 2024; 42:1283-1291. [PMID: 38310019 DOI: 10.1016/j.vaccine.2024.01.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/05/2024]
Abstract
Smallpox, caused by the variola virus belonging to the genus Orthopoxvirus, is an acute contagious disease that killed 300 million people in the 20th century. Since it was declared to be eradicated and the national immunization program against it was stopped, the variola virus has become a prospective bio-weapon. It is necessary to develop a safe vaccine that protects people from terrorism using this biological weapon and that can be administered to immunocompromised people. Our previous study reported on the development of an attenuated smallpox vaccine (KVAC103). This study evaluated cellular and humoral immune responses to various doses, frequencies, and routes of administration of the KVAC103 strain, compared to CJ-50300 vaccine, and its protective ability against the wild-type vaccinia virus Western Reserve (VACV-WR) strain was evaluated. The binding and neutralizing-antibody titers increased in a concentration-dependent manner in the second inoculation, which increased the neutralizing-antibody titer compared to those after the single injection. In contrast, the T-cell immune response (interferon-gamma positive cells) increased after the second inoculation compared to that of CJ-50300 after the first inoculation. Neutralizing-antibody titers and antigen-specific IgG levels were comparable in all groups administered KVAC103 intramuscularly, subcutaneously, and intradermally. In a protective immunity test using the VACV-WR strain, all mice vaccinated with CJ-50300 or KVAC103 showed 100% survival. KVAC103 could be a potent smallpox vaccine that efficiently induces humoral and cellular immune responses to protect mice against the VACV-WR strain.
Collapse
Affiliation(s)
- Yun-Ho Hwang
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu 28159, Republic of Korea
| | - Yeji Byeon
- BIO Research Institute, BIO-Pharmaceutical Research Center, 811 Deokpyeong-ro, Majang-myeon, Icheon-si, Gyeonggi-do 17389, Republic of Korea
| | - Sung Hyun Ahn
- BIO Research Institute, BIO-Pharmaceutical Research Center, 811 Deokpyeong-ro, Majang-myeon, Icheon-si, Gyeonggi-do 17389, Republic of Korea
| | - Mi-Young Kim
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu 28159, Republic of Korea
| | - Sung-Hyun Byun
- BIO Research Institute, BIO-Pharmaceutical Research Center, 811 Deokpyeong-ro, Majang-myeon, Icheon-si, Gyeonggi-do 17389, Republic of Korea
| | - Hyoung Jin Lee
- BIO Research Institute, BIO-Pharmaceutical Research Center, 811 Deokpyeong-ro, Majang-myeon, Icheon-si, Gyeonggi-do 17389, Republic of Korea
| | - Bohyun Suh
- BIO Research Institute, BIO-Pharmaceutical Research Center, 811 Deokpyeong-ro, Majang-myeon, Icheon-si, Gyeonggi-do 17389, Republic of Korea
| | - Dokeun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu 28159, Republic of Korea
| | - Eun Ju Jung
- BIO Research Institute, BIO-Pharmaceutical Research Center, 811 Deokpyeong-ro, Majang-myeon, Icheon-si, Gyeonggi-do 17389, Republic of Korea.
| | - You-Jin Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu 28159, Republic of Korea.
| |
Collapse
|
4
|
Thomas J, Garcia J, Terry M, Mahaney S, Quintanilla O, Silva DC, Morales M, VandeBerg JL. Monodelphis domestica as a Fetal Intra-Cerebral Inoculation Model for Zika Virus Pathogenesis. Pathogens 2023; 12:pathogens12050733. [PMID: 37242404 DOI: 10.3390/pathogens12050733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Monodelphis domestica (the laboratory opossum) is a marsupial native to South America. At birth, these animals are developmentally equivalent to human embryos at approximately 5 weeks of gestation, which, when coupled with other characteristics including the size of the animals, the development of a robust immune system during juvenile development, and the relative ease of experimental manipulation, have made M. domestica a valuable model in many areas of biomedical research. However, their suitability as models for infectious diseases, especially neurotropic viruses such as Zika virus (ZIKV), is currently unknown. Here, we describe the replicative effects of ZIKV using a fetal intra-cerebral model of inoculation. Using immunohistology and in situ hybridization, we found that opossum embryos and fetuses are susceptible to infection by ZIKV administered intra-cerebrally, that the infection persists, and that viral replication results in neural pathology and may occasionally result in global growth restriction. These results demonstrate the utility of M. domestica as a new animal model for investigating ZIKV infection in vivo and facilitate further inquiry into viral pathogenesis, particularly for those viruses that are neurotropic, that require a host with the ability to sustain sustained viremia, and/or that may require intra-cerebral inoculations of large numbers of embryos or fetuses.
Collapse
Affiliation(s)
- John Thomas
- Center for Vector Borne Disease, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- Department of Human Genetics, The University of Texas Rio Grande Valley, Brownsville, TX 78521, USA
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Juan Garcia
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Matthew Terry
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Susan Mahaney
- Department of Human Genetics, The University of Texas Rio Grande Valley, Brownsville, TX 78521, USA
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX 78521, USA
| | - Oscar Quintanilla
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Dionn Carlo Silva
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Marisol Morales
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - John L VandeBerg
- Center for Vector Borne Disease, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- Department of Human Genetics, The University of Texas Rio Grande Valley, Brownsville, TX 78521, USA
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX 78521, USA
| |
Collapse
|
5
|
Kushwaha V, Capalash N. Evaluation of immunomodulatory potential of recombinant histidyl-tRNA synthetase (rLdHisRS) protein of Leishmania donovani as a vaccine candidate against visceral leishmaniasis. Acta Trop 2023; 241:106867. [PMID: 36878386 DOI: 10.1016/j.actatropica.2023.106867] [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: 12/15/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 03/07/2023]
Abstract
Visceral leishmaniasis is neglected tropical protozoan disease caused by Leishmania donovani and are associated with high fatality rate in developing countries since prophylactic vaccines are not available. In the present study, we evaluated the immunomodulatory potential of L. donovani histidyl-tRNA synthetase (LdHisRS) and predicted the epitopes using immunoinformatic tools. Histidyl-tRNA synthetase (HisRS) is a class IIa aminoacyl t-RNA synthetase enzyme (aaRS) required for histidine incorporation into proteins during protein synthesis. The recombinant LdHisRS protein (rLdHisRS) was expressed in E coli BL-21cells, and its immunomodulatory role was assessed in J774A.1 murine macrophage and in BALB/c mice, respectively. LdHisRS specifically stimulated and triggered enhance cell proliferation, nitric oxide release and IFN-γ (70%; P < 0.001), and IL-12 (55.37%; P < 0.05) cytokine release in vitro, whereas BALB/c mice immunized with rLdHisRS show higher NO release (80.95%; P<0.001), higher levels of Th1 cytokines IFN-γ (14%; P < 0.05), TNF-α (34.93%; P < 0.001), and IL-12 (28.49%; P < 0.001) and robust IgG (p<0.001) and IgG2a (p<0.001) production. We also identified 20 Helper T-lymphocytes (HTLs), 30 cytotoxic T lymphocytes (CTLs), and 18 B-cell epitopes from HisRS protein of L. donovani. All these epitopes can be further used to make a multiepitope vaccine against L. donovani.
Collapse
Affiliation(s)
- Vikas Kushwaha
- Department of Biotechnology, Panjab University, Sector-25, South Campus, Chandigarh 160025, India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Sector-25, South Campus, Chandigarh 160025, India.
| |
Collapse
|
6
|
León B. Understanding the development of Th2 cell-driven allergic airway disease in early life. FRONTIERS IN ALLERGY 2023; 3:1080153. [PMID: 36704753 PMCID: PMC9872036 DOI: 10.3389/falgy.2022.1080153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Allergic diseases, including atopic dermatitis, allergic rhinitis, asthma, and food allergy, are caused by abnormal responses to relatively harmless foreign proteins called allergens found in pollen, fungal spores, house dust mites (HDM), animal dander, or certain foods. In particular, the activation of allergen-specific helper T cells towards a type 2 (Th2) phenotype during the first encounters with the allergen, also known as the sensitization phase, is the leading cause of the subsequent development of allergic disease. Infants and children are especially prone to developing Th2 cell responses after initial contact with allergens. But in addition, the rates of allergic sensitization and the development of allergic diseases among children are increasing in the industrialized world and have been associated with living in urban settings. Particularly for respiratory allergies, greater susceptibility to developing allergic Th2 cell responses has been shown in children living in urban environments containing low levels of microbial contaminants, principally bacterial endotoxins [lipopolysaccharide (LPS)], in the causative aeroallergens. This review highlights the current understanding of the factors that balance Th2 cell immunity to environmental allergens, with a particular focus on the determinants that program conventional dendritic cells (cDCs) toward or away from a Th2 stimulatory function. In this context, it discusses transcription factor-guided functional specialization of type-2 cDCs (cDC2s) and how the integration of signals derived from the environment drives this process. In addition, it analyzes observational and mechanistic studies supporting an essential role for innate sensing of microbial-derived products contained in aeroallergens in modulating allergic Th2 cell immune responses. Finally, this review examines whether hyporesponsiveness to microbial stimulation, particularly to LPS, is a risk factor for the induction of Th2 cell responses and allergic sensitization during infancy and early childhood and the potential factors that may affect early-age response to LPS and other environmental microbial components.
Collapse
Affiliation(s)
- Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
7
|
Sun Y, Hu B, Stanley G, Harris ZM, Gautam S, Homer R, Koff JL, Rajagopalan G. IFN- γ Is Protective in Cytokine Release Syndrome-associated Extrapulmonary Acute Lung Injury. Am J Respir Cell Mol Biol 2023; 68:75-89. [PMID: 36125351 PMCID: PMC9817908 DOI: 10.1165/rcmb.2022-0117oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/19/2022] [Indexed: 02/05/2023] Open
Abstract
The mechanisms by which excessive systemic activation of adaptive T lymphocytes, as in cytokine release syndrome (CRS), leads to innate immune cell-mediated acute lung injury (ALI) or acute respiratory distress syndrome, often in the absence of any infection, remains unknown. Here, we investigated the roles of IFN-γ and IL-17A, key T-cell cytokines significantly elevated in patients with CRS, in the immunopathogenesis of CRS-induced extrapulmonary ALI. CRS was induced in wild-type (WT), IL-17A- and IFN-γ knockout (KO) human leukocyte antigen-DR3 transgenic mice with 10 μg of the superantigen, staphylococcal enterotoxin B, given intraperitoneally. Several ALI parameters, including gene expression profiling in the lungs, were studied 4, 24, or 48 hours later. Systemic T-cell activation with staphylococcal enterotoxin B resulted in robust upregulation of several chemokines, S100A8/A9, matrix metalloproteases, and other molecules implicated in tissue damage, granulocyte as well as agranulocyte adhesion, and diapedesis in the lungs as early as 4 hours, which was accompanied by subsequent neutrophil/eosinophil lung infiltration and severe ALI in IFN-γ KO mice. These pathways were significantly underexpressed in IL-17A KO mice, which manifested mildest ALI and intermediate in WT mice. Neutralization of IFN-γ worsened ALI in WT and IL-17A KO mice, whereas neutralizing IL-17A did not mitigate lung injury in IFN-γ KO mice, suggesting a dominant protective role for IFN-γ in ALI and that IL-17A is dispensable. Ruxolitinib, a Janus kinase inhibitor, increased ALI severity in WT mice. Thus, our study identified novel mechanisms of ALI in CRS and its differential modulation by IFN-γ and IL-17A.
Collapse
Affiliation(s)
- Ying Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Buqu Hu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Gail Stanley
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Zachary M. Harris
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Samir Gautam
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Robert Homer
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; and
- Pathology and Laboratory Medicine Service, Veterans Affairs Connecticut HealthCare System, West Haven, Connecticut
| | - Jonathan L. Koff
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | | |
Collapse
|
8
|
Conde L, Maciel G, de Assis GM, Freire-de-Lima L, Nico D, Vale A, Freire-de-Lima CG, Morrot A. Humoral response in Leishmaniasis. Front Cell Infect Microbiol 2022; 12:1063291. [PMID: 36579347 PMCID: PMC9791258 DOI: 10.3389/fcimb.2022.1063291] [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: 10/06/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022] Open
Abstract
Leishmaniasis presents different types of clinical manifestations that can be divided into cutaneous leishmaniasis and visceral leishmaniasis. The host's immune system, associated with genetic and nutritional factors, is strongly involved in the evolution of the disease or parasite escape. Humoral immunity is characterized by the production of antibodies capable of promoting neutralization, opsonization, and activation of the complement system. In this scenario, B lymphocytes produce antibodies that play an important role in Leishmania infection although neglected for a long time. Thus, relevant aspects in the establishment of Leishmania infection will be addressed, highlighting the importance of humoral immunity during the entire process of Leishmania infection.
Collapse
Affiliation(s)
- Luciana Conde
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriela Maciel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Meira de Assis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dirlei Nico
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Vale
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Alexandre Morrot
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil,*Correspondence: Alexandre Morrot,
| |
Collapse
|
9
|
Gurjar D, Kumar Patra S, Bodhale N, Lenka N, Saha B. Leishmania intercepts IFN-γR signaling at multiple levels in macrophages. Cytokine 2022; 157:155956. [PMID: 35785668 DOI: 10.1016/j.cyto.2022.155956] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022]
Abstract
IFN-γ, a type 2 interferon and a cytokine, is critical for both innate and adaptive immunity. IFN-γ binds to the IFN-γRs on the cell membrane of macrophages, signals through JAK1-STAT-1 pathway and induces IFN-γ-stimulated genes (ISGs). As Leishmania amastigotes reside and replicate within macrophages, IFN-γ mediated macrophage activation eventuate in Leishmania elimination. As befits the principle of parasitism, the impaired IFN-γ responsiveness in macrophages ensures Leishmania survival. IFN-γ responsiveness is a function of integrated molecular events at multiple levels in the cells that express IFN-γ receptors. In Leishmania-infected macrophages, reduced IFN-γRα expression, impaired IFN-γRα and IFN-γRβ hetero-dimerization due to altered membrane lipid composition, reduced JAK-1 and STAT-1 phosphorylation but increased STAT-1 degradation and impaired ISGs induction collectively determine the IFN-γ responsiveness and the efficacy of IFN-γ induced antileishmanial function of macrophages. Therefore, parasite load is not only decided by the levels of IFN-γ produced but also by the IFN-γ responsiveness. Indeed, in Leishmania-infected patients, IFN-γ is produced but IFN-γ signalling is downregulated. However, the molecular mechanisms of IFN-γ responsiveness remain unclear. Therefore, we review the current understanding of IFN-γ responsiveness of Leishmania-infected macrophages.
Collapse
Affiliation(s)
- Dhiraj Gurjar
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | | | - Neelam Bodhale
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Nibedita Lenka
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| |
Collapse
|
10
|
Machado AS, Lage DP, Vale DL, Freitas CS, Linhares FP, Cardoso JMO, Oliveira-da-Silva JA, Pereira IAG, Ramos FF, Tavares GSV, Ludolf F, Bandeira RS, Maia LGN, Menezes-Souza D, Duarte MC, Chávez-Fumagalli MA, Roatt BM, Christodoulides M, Martins VT, Coelho EAF. Leishmania LiHyC protein is immunogenic and induces protection against visceral leishmaniasis. Parasite Immunol 2022; 44:e12921. [PMID: 35437797 DOI: 10.1111/pim.12921] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 11/27/2022]
Abstract
AIMS Treatment against visceral leishmaniasis (VL) presents problems by toxicity of drugs, high cost and/or emergence of resistant strains. The diagnosis is hampered by variable sensitivity and/or specificity of tests. In this context, prophylactic vaccination could represent a control measure against disease. In this study, the protective efficacy from Leishmania LiHyC protein was evaluated in murine model against Leishmania infantum infection. METHODS AND RESULTS LiHyC was used as recombinant protein (rLiHyC) associated with saponin (rLiHyC/S) or Poloxamer 407-based polymeric micelles (rLiHyC/M) to immunize mice. Animals received also saline, saponin or empty micelles as controls. The immunogenicity was evaluated before and after challenge, and results showed that vaccination with rLiHyC/S or rLiHyC/M induced the production of high levels of IFN-γ, IL-12 and GM-CSF in cell culture supernatants, as well as higher IFN-γ expression evaluated by RT-qPCR and involvement from CD4+ and CD8+ T cell subtypes producing IFN-γ, TNF-α and IL-2. A positive lymphoproliferative response was also found in cell cultures from vaccinated animals, besides high levels of rLiHyC- and parasite-specific nitrite and IgG2a antibodies. Immunological assays correlated with significant reductions in the parasite load in spleens, livers, bone marrows and draining lymph nodes from vaccinated mice, when compared to values found in the controls. The micellar composition showed slightly better immunological and parasitological data, as compared to rLiHyC/S. CONCLUSION Results suggest that rLiHyC associated with adjuvants could be considered for future studies as a vaccine candidate against VL.
Collapse
Affiliation(s)
- Amanda S Machado
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela P Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Danniele L Vale
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Camila S Freitas
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Flávia P Linhares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Jamille M O Cardoso
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - João A Oliveira-da-Silva
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Isabela A G Pereira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda F Ramos
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Ludolf
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Raquel S Bandeira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz G N Maia
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Menezes-Souza
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil.,Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana C Duarte
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil.,Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Miguel A Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José S/N, Umacollo, Arequipa, Peru
| | - Bruno M Roatt
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Myron Christodoulides
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, England
| | - Vívian T Martins
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil.,Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
11
|
Karasarides M, Cogdill AP, Robbins PB, Bowden M, Burton EM, Butterfield LH, Cesano A, Hammer C, Haymaker CL, Horak CE, McGee HM, Monette A, Rudqvist NP, Spencer CN, Sweis RF, Vincent BG, Wennerberg E, Yuan J, Zappasodi R, Lucey VMH, Wells DK, LaVallee T. Hallmarks of Resistance to Immune-Checkpoint Inhibitors. Cancer Immunol Res 2022; 10:372-383. [PMID: 35362046 PMCID: PMC9381103 DOI: 10.1158/2326-6066.cir-20-0586] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/15/2021] [Accepted: 01/24/2022] [Indexed: 01/29/2023]
Abstract
Immune-checkpoint inhibitors (ICI), although revolutionary in improving long-term survival outcomes, are mostly effective in patients with immune-responsive tumors. Most patients with cancer either do not respond to ICIs at all or experience disease progression after an initial period of response. Treatment resistance to ICIs remains a major challenge and defines the biggest unmet medical need in oncology worldwide. In a collaborative workshop, thought leaders from academic, biopharma, and nonprofit sectors convened to outline a resistance framework to support and guide future immune-resistance research. Here, we explore the initial part of our effort by collating seminal discoveries through the lens of known biological processes. We highlight eight biological processes and refer to them as immune resistance nodes. We examine the seminal discoveries that define each immune resistance node and pose critical questions, which, if answered, would greatly expand our notion of immune resistance. Ultimately, the expansion and application of this work calls for the integration of multiomic high-dimensional analyses from patient-level data to produce a map of resistance phenotypes that can be utilized to guide effective drug development and improved patient outcomes.
Collapse
Affiliation(s)
- Maria Karasarides
- Worldwide Medical Oncology, Bristol Myers Squibb, Princeton, New Jersey
| | - Alexandria P. Cogdill
- Immunai, New York, New York
- Department of Immunology, The University of Texas MD Anderson, Houston, Texas
| | | | - Michaela Bowden
- Translational Medicine, Bristol Myers Squibb, Cambridge, Massachusetts
| | - Elizabeth M. Burton
- Department of Surgical Oncology, The University of Texas MD Anderson, Houston, Texas
| | - Lisa H. Butterfield
- Parker Institute for Cancer Immunotherapy, San Francisco, California
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California
| | | | - Christian Hammer
- Department of Cancer Immunology, Genentech, South San Francisco, California
- Department of Human Genetics, Genentech, South San Francisco, California
| | - Cara L. Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christine E. Horak
- Global Drug Development, Bristol Myers Squibb, Lawrenceville, New Jersey
| | - Heather M. McGee
- Department of Radiation Oncology, City of Hope National Medical Center and Department of Immuno-Oncology, Beckmann Research Institute, City of Hope, Duarte, California
| | - Anne Monette
- Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | | | - Christine N. Spencer
- Department of Informatics, Parker Institute for Cancer Immunotherapy, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Randy F. Sweis
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
- Committee on Immunology, University of Chicago, Chicago, Illinois
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | | | - Jianda Yuan
- Translational Oncology, Early Oncology Development Department, Merck Research Laboratories, Rahway, New Jersey
| | - Roberta Zappasodi
- Weill Cornell Medicine, New York, New York
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Daniel K. Wells
- Immunai, New York, New York
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Theresa LaVallee
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| |
Collapse
|
12
|
Montakhab-Yeganeh H, Shafiei R, Najm M, Masoori L, Aspatwar A, Badirzadeh A. Immunogenic properties of empty pcDNA3 plasmid against zoonotic cutaneous leishmaniasis in mice. PLoS One 2022; 17:e0263993. [PMID: 35167596 PMCID: PMC8846536 DOI: 10.1371/journal.pone.0263993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
Background Leishmania (L) parasite, the causative agent of zoonotic cutaneous leishmaniasis (ZCL), effectively stimulates the mammalian cells to mount strong humoral responses by enhancing T-helper-2 (Th2)-associated cytokines for its survival. The best strategy to decrease the intensity of infection in the host is induction of cellular immunity. Methods We evaluated the effects of the empty bacterial pcDNA3 plasmid on mice infected with L. major and quantified the immune mediators including IFN-γ, IL-4, IL-10, IgG2a, IgG1, arginase activity and nitric oxide (NO) in the mice. Moreover, the footpad lesion size and parasite load were assessed. Results We observed that pcDNA3 could modulate the immune responses in favor of host cells and decrease the disease severity. Th2- associated mediators, including arginase, IL-4, and IL-10 are downregulated, while cellular responses are upregulated in line with an increase in the levels of nitric oxide (NO) and interfero-gamma (IFN-γ). Interestingly, pcDNA3 induced specific Th1-associated antibodies, IgG2a isotype; however, it suppressed the production of humoral IgG1. The stimulation of the immune response by the empty pcDNA3 is able to shift the immune function to predominant cellular responses caused by Th1, and it had a positive effect on the treatment of zoonotic cutaneous leishmaniasis (ZCL). Conclusions Altogether, we introduced the pcDNA3 as a potential interfering factor in the modulation of the immune system against ZCL. Since this vector has been widely used as a control group in different studies, we suggest that the potential function of the empty vector should be deeply assessed, as it exerts anti-parasitic effects on mice infected with L. major.
Collapse
Affiliation(s)
- Hossein Montakhab-Yeganeh
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Reza Shafiei
- Vector-borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Najm
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Masoori
- Department of Laboratory Sciences, School of Allied Medical Sciences, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alireza Badirzadeh
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- * E-mail: ,
| |
Collapse
|
13
|
Lopes ME, dos Santos LM, Sacks D, Vieira LQ, Carneiro MB. Resistance Against Leishmania major Infection Depends on Microbiota-Guided Macrophage Activation. Front Immunol 2021; 12:730437. [PMID: 34745100 PMCID: PMC8564857 DOI: 10.3389/fimmu.2021.730437] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
Innate immune cells present a dual role during leishmaniasis: they constitute the first line of host defense but are also the main host cells for the parasite. Response against the infection that results in the control of parasite growth and lesion healing depends on activation of macrophages into a classical activated phenotype. We report an essential role for the microbiota in driving macrophage and monocyte-derived macrophage activation towards a resistance phenotype against Leishmania major infection in mice. Both germ-free and dysbiotic mice showed a higher number of myeloid innate cells in lesions and increased number of infected cells, mainly dermal resident and inflammatory macrophages. Despite developing a Th1 immune response characterized by the same levels of IFN-γ production as the conventional mice, germ-free mice presented reduced numbers of iNOS+ macrophages at the peak of infection. Absence or disturbance of host microbiota impaired the capacity of bone marrow-derived macrophage to be activated for Leishmania killing in vitro, even when stimulated by Th1 cytokines. These cells presented reduced expression of inos mRNA, and diminished production of microbicidal molecules, such as ROS, while presenting a permissive activation status, characterized by increased expression of arginase I and il-10 mRNA and higher arginase activity. Colonization of germ-free mice with complete microbiota from conventional mice rescued their ability to control the infection. This study demonstrates the essential role of host microbiota on innate immune response against L. major infection, driving host macrophages to a resistance phenotype.
Collapse
Affiliation(s)
- Mateus Eustáquio Lopes
- Laboratório de Gnotobiologia e Imunologia, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Liliane Martins dos Santos
- Laboratório de Gnotobiologia e Imunologia, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - David Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Leda Quercia Vieira
- Laboratório de Gnotobiologia e Imunologia, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus B. Carneiro
- Laboratório de Gnotobiologia e Imunologia, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
14
|
Tomiotto-Pellissier F, Miranda-Sapla MM, Silva TF, Bortoleti BTDS, Gonçalves MD, Concato VM, Rodrigues ACJ, Detoni MB, Costa IN, Panis C, Conchon-Costa I, Bordignon J, Pavanelli WR. Murine Susceptibility to Leishmania amazonensis Infection Is Influenced by Arginase-1 and Macrophages at the Lesion Site. Front Cell Infect Microbiol 2021; 11:687633. [PMID: 34660334 PMCID: PMC8517480 DOI: 10.3389/fcimb.2021.687633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/13/2021] [Indexed: 12/30/2022] Open
Abstract
Cutaneous leishmaniasis is a zoonotic infectious disease broadly distributed worldwide, causing a range of diseases with clinical outcomes ranging from self-healing infections to chronic disfiguring disease. The effective immune response to this infection is yet to be more comprehensively understood and is fundamental for developing drugs and vaccines. Thus, we used experimental models of susceptibility (BALB/c) and partial resistance (C57BL/6) to Leishmania amazonensis infection to investigate the local profile of mediators involved in the development of cutaneous leishmaniasis. We found worse disease outcome in BALB/c mice than in C57BL/6 mice, with almost 15 times higher parasitic load, ulcerated lesion formation, and higher levels of IL-6 in infected paws. In contrast, C57BL/6 presented higher levels of IFN-γ and superoxide anion (•O2−) after 11 weeks of infection and no lesion ulcerations. A peak of local macrophages appeared after 24 h of infection in both of the studied mice strains, followed by another increase after 240 h, detected only in C57BL/6 mice. Regarding M1 and M2 macrophage phenotype markers [iNOS, MHC-II, CD206, and arginase-1 (Arg-1)], we found a pronounced increase in Arg-1 levels in BALB/c after 11 weeks of infection, whereas C57BL/6 showed an initial predomination of markers from both profiles, followed by an M2 predominance, coinciding with the second peak of macrophage infiltration, 240 h after the infection. Greater deposition of type III collagen and lesion resolution was also observed in C57BL/6 mice. The adoptive transfer of macrophages from C57BL/6 to infected BALB/c at the 11th week showed a reduction in both edema and the number of parasites at the lesion site, in addition to lower levels of Arg-1. Thus, C57BL/6 mice have a more effective response against L. amazonensis, based on a balance between inflammation and tissue repair, while BALB/c mice have an excessive Arg-1 production at late infection. The worst evolution seems to be influenced by recruitment of Arg-1 related macrophages, since the adoptive transfer of macrophages from C57BL/6 mice to BALB/c resulted in better outcomes, with lower levels of Arg-1.
Collapse
Affiliation(s)
- Fernanda Tomiotto-Pellissier
- Biosciences and Biotechnology Graduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Milena Menegazzo Miranda-Sapla
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Taylon Felipe Silva
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Bruna Taciane da Silva Bortoleti
- Biosciences and Biotechnology Graduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Manoela Daiele Gonçalves
- Laboratory of Biotransformation and Phytochemistry, Department of Chemistry, State University of Londrina, Universitary Hospital, Londrina, Brazil
| | - Virginia Márcia Concato
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Ana Carolina Jacob Rodrigues
- Biosciences and Biotechnology Graduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Mariana Barbosa Detoni
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Idessania Nazareth Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of Western Paraná (UNIOESTE), Francisco Beltrão, Brazil
| | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Juliano Bordignon
- Biosciences and Biotechnology Graduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Molecular Virology, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil
| | - Wander Rogério Pavanelli
- Biosciences and Biotechnology Graduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology of Neglected Diseases and Cancer (LIDNC), Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
15
|
da Silva GAV, Mesquita TG, Souza VC, Junior JDES, Gomes de Souza ML, Talhari AC, Talhari S, Naveca FG, Ramasawmy R. A Single Haplotype of IFNG Correlating With Low Circulating Levels of Interferon-γ Is Associated With Susceptibility to Cutaneous Leishmaniasis Caused by Leishmania guyanensis. Clin Infect Dis 2021; 71:274-281. [PMID: 31722386 DOI: 10.1093/cid/ciz810] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/22/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Interferon-γ (IFN-γ) plays an important role in the control of Leishmania infection. Blockade of IFN-γ signaling in mice increases lesion size and parasite load. In endemic areas of Leishmaniasis, only a fraction of the population develop the disease. This suggest that host genetics may play a role in this response. We investigated whether single nucleotide polymorphisms (SNPs) in IFNG may be associated with elevated or decrease risk in the development of cutaneous leishmaniasis (CL). METHODS We assessed 9 SNP and cytosine-adenine (CA) repeats in IFNG by nucleotide sequencing in 647 patients with CL caused by Leishmania guyanensis and 629 controls. Circulating plasma IFN-γ levels were also assayed in 400 patients with CL and 400 controls. RESULTS The rs2069705TT genotype is associated with elevated risk of developing CL compared with the rs2069705CC genotype (OR, 1.7; 95% CI, 1.3-2.4; P = .0008). There is a 70% chance that this genotype raises the risk of developing CL. In a dominant model, carriers of the rs2069705T allele compared with the rs2069705CC genotype showed a 50% (range, 20-100%) increased risk of developing CL (OR, 1.5; 95% CI, 1.2-2.0; P = .0004). Haplotype analysis showed 1 haplotype (H1) associated with low levels of IFN-γ presented an increased risk of 60% of developing CL (OR, 1.6; 95% CI, 1.3-1.9; P = 5 × 10-5) compared with non-H1. CONCLUSIONS IFNG variant rs2069705 seems to be a genetic modifier of clinical outcome of Leishmania infection; individuals with the H1 haplotype, associated with low levels of IFN-γ, have a 60% risk of developing CL.
Collapse
Affiliation(s)
- George A V da Silva
- Programa de Pos-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Amazonas, Manaus, Brazil
| | - Tirza G Mesquita
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Amazonas, Manaus, Brazil
| | - Victor C Souza
- Instituto Leônidas e Maria Deane, FIOCRUZ Amazônia, Amazonas, Manaus, Brazil
| | - José do Espírito Santo Junior
- Programa de Pos-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Amazonas, Manaus, Brazil
| | | | | | - Sinésio Talhari
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Amazonas, Brazil.,Faculdade de Medicina, Universidade Nilton Lins, Amazonas, Manaus, Brazil
| | - Felipe G Naveca
- Instituto Leônidas e Maria Deane, FIOCRUZ Amazônia, Amazonas, Manaus, Brazil
| | - Rajendranath Ramasawmy
- Programa de Pos-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Amazonas, Manaus, Brazil.,Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Amazonas, Brazil.,Faculdade de Medicina, Universidade Nilton Lins, Amazonas, Manaus, Brazil
| |
Collapse
|
16
|
Chauhan P, Nair A, Patidar A, Dandapat J, Sarkar A, Saha B. A primer on cytokines. Cytokine 2021; 145:155458. [PMID: 33581983 DOI: 10.1016/j.cyto.2021.155458] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/19/2022]
Abstract
Cytokines are pleiotropic polypeptides that control the development of and responses mediated by immune cells. Cytokine classification predominantly relies on [1] the target receptor(s), [2] the primary structural features of the extracellular domains of their receptors, and [3] their receptor composition. Functionally, cytokines are either pro-inflammatory or anti-inflammatory, hematopoietic colony-stimulating factors, developmental and would healing maintaining immune homeostasis. When the balance in C can form complex networks amongst themselves that may affect the homeostasis and diseases. Cytokines can affect resistance and susceptibility for many diseases and their availability in the host cytokine production and interaction is disturbed, immunopathogenesis sets in. Therefore, cytokine-targeting bispecific, and chimeric antibodies form a significant mode of immnuo-therapeutics Although the field has grown deep and wide, many areas of cytokine biology remain unknown. Here, we have reviewed these cytokines along with the organization, signaling, and functions through respective cytokine-receptor-families. Being part of the special issue on the Role of Cytokines in Leishmaniasis, this review is intended to be used as an organized primer on cytokines and not a resource for detailed discussion- for which a two-volume Handbook of cytokines is available- on each of the cytokines. Priming the readers on cytokines, we next brief the role of cytokines in Leishmaniasis. In the brief, we do not provide an account of each of the involved cytokines known to date, instead, we offer a temporal relationship between the cytokines and the progress of the infection towards the alternate outcomes- healing or non-healing- of the infection.
Collapse
Affiliation(s)
- Prashant Chauhan
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Arathi Nair
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Ashok Patidar
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Jagneshwar Dandapat
- P.G. Department of Biotechnology, Utkal University, Bhubaneswar 751004, India
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar 751024, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India; Trident Academy of Creative Technology, Bhubaneswar 751024, India; Department of Allied Health Sciences, BLDE (Deemed University), Vijayapura 562135, India.
| |
Collapse
|
17
|
Yang Z, Wu CAM, Targ S, Allen CDC. IL-21 is a broad negative regulator of IgE class switch recombination in mouse and human B cells. J Exp Med 2020; 217:133860. [PMID: 32130409 PMCID: PMC7201927 DOI: 10.1084/jem.20190472] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 11/24/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
IgE antibodies may elicit potent allergic reactions, and their production is tightly controlled. The tendency to generate IgE has been thought to reflect the balance between type 1 and type 2 cytokines, with the latter promoting IgE. Here, we reevaluated this paradigm by a direct cellular analysis, demonstrating that IgE production was not limited to type 2 immune responses yet was generally constrained in vivo. IL-21 was a critical negative regulator of IgE responses, whereas IFN-γ, IL-6, and IL-10 were dispensable. Follicular helper T cells were the primary source of IL-21 that inhibited IgE responses by directly engaging the IL-21 receptor on B cells and triggering STAT3-dependent signaling. We reconciled previous discordant results between mouse and human B cells and revealed that the inhibition of IgE class switch recombination by IL-21 was attenuated by CD40 signaling, whereas IgG1 class switch recombination was potentiated by IL-21 in the context of limited IL-4. These findings establish key features of the extrinsic regulation of IgE production by cytokines.
Collapse
Affiliation(s)
- Zhiyong Yang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Chung-An M Wu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Sasha Targ
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA.,Department of Anatomy, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
18
|
Poudel B, Yorek MS, Mazgaeen L, Brown SA, Kanneganti TD, Gurung P. Acute IL-4 Governs Pathogenic T Cell Responses during Leishmania major Infection. Immunohorizons 2020; 4:546-560. [PMID: 32948646 PMCID: PMC7640617 DOI: 10.4049/immunohorizons.2000076] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/01/2020] [Indexed: 11/19/2022] Open
Abstract
Leishmania spp. infection is a global health problem affecting more than 2 million people every year with 300 million at risk worldwide. It is well established that a dominant Th1 response (IFN-γ, a hallmark Th1 cytokine) provides resistance, whereas a dominant Th2 response (IL-4, a hallmark Th2 cytokine) confers susceptibility during infection. Given the important role of IL-4 during L. major infection, we used IL-4-neutralizing Abs to investigate the cellular and molecular events regulated by IL-4 signaling. As previously published, neutralization of IL-4 in L. major-infected BALB/c mice (a Leishmania susceptible strain) provided protection when compared with control L. major-infected BALB/c mice. Despite this protection, IFN-γ production by T cells was dramatically reduced. Temporal neutralization of IL-4 revealed that acute IL-4 produced within the first days of infection is critical for not only programming IL-4-producing Th2 CD4+ T cells, but for promoting IFN-γ produced by CD8+ T cells. Mechanistically, IL-4 signaling enhances anti-CD3-induced Tbet and IFN-γ expression in both CD4+ and CD8+ T cells. Given the pathogenic role of IFN-γ-producing CD8+ T cells, our data suggest that IL-4 promotes cutaneous leishmaniasis pathology by not only promoting Th2 immune responses but also pathogenic CD8+ T cell responses. Our studies open new research grounds to investigate the unsuspected role of IL-4 in regulating both Th1 and Th2 responses.
Collapse
Affiliation(s)
- Barun Poudel
- Iowa Inflammation Program, University of Iowa, Iowa City, IA 52242.,Department of Internal Medicine, University of Iowa, Iowa City, IA 52242
| | - Matthew S Yorek
- Iowa Inflammation Program, University of Iowa, Iowa City, IA 52242.,Department of Internal Medicine, University of Iowa, Iowa City, IA 52242
| | - Lalita Mazgaeen
- Iowa Inflammation Program, University of Iowa, Iowa City, IA 52242.,Department of Internal Medicine, University of Iowa, Iowa City, IA 52242.,Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA 52242
| | - Scott A Brown
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | - Prajwal Gurung
- Iowa Inflammation Program, University of Iowa, Iowa City, IA 52242; .,Department of Internal Medicine, University of Iowa, Iowa City, IA 52242.,Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA 52242.,Immunology Graduate Program, University of Iowa, Iowa City, IA 52242; and.,Center for Immunology and Immune Based Disease, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
19
|
de Araújo-Souza PS, Hanschke SCH, Nardy AFFR, Sécca C, Oliveira-Vieira B, Silva KL, Soares-Lima SC, Viola JPB. Differential interferon-γ production by naive and memory-like CD8 T cells. J Leukoc Biol 2020; 108:1329-1337. [PMID: 32421902 DOI: 10.1002/jlb.2ab0420-646r] [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: 12/16/2019] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 11/08/2022] Open
Abstract
CD8 T cells play a crucial role in immune responses to virus infections and tumors. Naïve CD8 T lymphocytes after TCR stimulation undergo differentiation into CTLs and memory cells, which are essential sources of IFN-γ. We investigated IFN-γ production by CD8 T cell subsets found in nonimmune mice. A minor fraction of in vitro TCR-stimulated CD8 T cells produce IFN-γ, and it is regulated at the transcriptional level. Antigen inexperienced C57BL/6 mice present the coexistence of 2 populations. The main population exhibits a CD44low CD122low profile, which is compatible with naïve lymphocytes. The minor expresses a phenotype of immunologic memory, CD44hi CD122hi . Both subsets are able to produce IL-2 in response to TCR activation, but only the memory-like population is responsible for IFN-γ production. Similar to memory CD8 T cells, CD44hi CD8+ T cells also present a higher level of the transcriptional factor Eomes and a lower level of T-bet (Tbx21) mRNA than CD44low CD8+ T cells. The presence of the CD44hi CD8+ T cell population in nonimmune OT-I transgenic mice reveals that the population is generated independently of antigenic stimulation. CpG methylation is an efficient epigenetic mechanism for gene silencing. DNA methylation at posttranscriptional CpG sites in the Ifng promoter is higher in CD44low CD8+ T cells than in CD44hi CD8+ T cells. Thus, memory-like CD8 T cells have a distinct epigenetic pattern in the Ifng promoter and can rapidly produce IFN-γ in response to TCR stimulation.
Collapse
Affiliation(s)
- Patrícia S de Araújo-Souza
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil.,Department of Immunobiology, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Steffi C H Hanschke
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Ana Flavia F R Nardy
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Cristiane Sécca
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Barbara Oliveira-Vieira
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Karina L Silva
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Sheila C Soares-Lima
- Program of Molecular Carcinogenesis, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - João P B Viola
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
20
|
Ishizuka K, Fujii W, Azuma N, Mizobuchi H, Morimoto A, Sanjoba C, Matsumoto Y, Goto Y. Pathological roles of MRP14 in anemia and splenomegaly during experimental visceral leishmaniasis. PLoS Negl Trop Dis 2020; 14:e0008020. [PMID: 31961866 PMCID: PMC6994150 DOI: 10.1371/journal.pntd.0008020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/31/2020] [Accepted: 01/01/2020] [Indexed: 01/03/2023] Open
Abstract
Myeloid-related protein 14 (MRP14) belongs to the S100 calcium-binding protein family and is expressed in neutrophils and inflammatory macrophages. Increase in the number of MRP14+ cells or serum level of MRP14 is associated with various diseases such as autoimmune diseases and infectious diseases, suggesting the involvement of the molecule in pathogenesis of those diseases. In this study, to examine the pathological involvement of MRP14 during cutaneous and visceral leishmaniasis, wild-type (WT) and MRP14 knockout (MRP14KO) mice were infected with Leishmania major and L. donovani. Increase in the number of MRP14+ cells at the infection sites in wild-type mice was commonly found in the skin during L. major infection as well as the spleen and liver during L. donovani infection. In contrast, the influence of MRP14 to the pathology seemed different between the two infections. MRP14 depletion exacerbated the lesion development and ulcer formation in L. major infection. On the other hand, the depletion improved anemia and splenomegaly but not hepatomegaly at 24 weeks of L. donovani infection. These results suggest that, distinct from its protective role in CL, MRP14 is involved in exacerbation of some symptoms during VL.
Collapse
Affiliation(s)
- Kanna Ishizuka
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Wataru Fujii
- Laboratory of Applied Genetics, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Natsuho Azuma
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Haruka Mizobuchi
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ayako Morimoto
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Chizu Sanjoba
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshitsugu Matsumoto
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Goto
- Laboratory of Molecular Immunology, Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
21
|
A new multi-epitope peptide vaccine induces immune responses and protection against Leishmania infantum in BALB/c mice. Med Microbiol Immunol 2019; 209:69-79. [PMID: 31696313 DOI: 10.1007/s00430-019-00640-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023]
Abstract
Visceral leishmaniasis (VL) is a tropical and subtropical disease which is endemic in more than eighty countries around the world. Leishmania infantum is one of the main causative agents of VL disease. Currently, there is no approved-to-market vaccine for VL therapy. In this study, we evaluated cellular and humoral immune responses induced by our newly designed multi-epitope vaccine in BALB/c mice. Four antigenic proteins, including histone H1, sterol 24-c-methyltransferase (SMT), Leishmania-specific hypothetical protein (LiHy), and Leishmania-specific antigenic protein (LSAP) were chosen for the prediction of potential immunodominant epitopes. Moreover, to enhance vaccine immunogenicity, two toll-like receptors 4 (TLR4) agonists, resuscitation-promoting factors of Mycobacterium tuberculosis (RpfE and RpfB), were employed as the built-in adjuvants. Immunization with the designed multi-epitope vaccine elicited a robust Th1-type immune response, compared to other groups, as shown by increased levels of IL-2, IFN-γ, TNF-α, and IgG2a. Furthermore, a significant decrease was observed in Th-2-type-related cytokines such as IL-4 in immunized mice. The designed construct also induced a significant reduction in parasite load (p < 0.0001), conferring protection against L. infantum challenge. This study could be promising in gaining insight towards the potential of peptide epitope-based vaccines as effective protective approaches against Leishmania species.
Collapse
|
22
|
Zahid MSH, Johnson MM, Tokarski RJ, Satoskar AR, Fuchs JR, Bachelder EM, Ainslie KM. Evaluation of synergy between host and pathogen-directed therapies against intracellular Leishmania donovani. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2019; 10:125-132. [PMID: 31493763 PMCID: PMC6731340 DOI: 10.1016/j.ijpddr.2019.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 11/24/2022]
Abstract
Visceral leishmaniasis (VL) is associated with treatment complications due to the continued growth of resistant parasites toward currently available pathogen-directed therapeutics. To limit the emergence and combat resistant parasites there is a need to develop new anti-leishmanial drugs and alternative treatment approaches, such as host-directed therapeutics (HDTs). Discovery of new anti-leishmanial drugs including HDTs requires suitable in vitro assay systems. Herein, we modified and evaluated a series of resazurin assays against different life-stages of the VL causing parasite, Leishmania donovani to identify novel HDTs. We further analyzed the synergy of combinatorial interactions between traditionally used pathogen-directed drugs and HDTs for clearance of intracellular L. donovani. The inhibitory concentration at 50% (IC50) of the five evaluated therapies [amphotericin B (AMB), miltefosine, paromomycin, DNER-4, and AR-12 (OSU-03012)] was determined against promastigotes, extracellular amastigotes, and intracellular amastigotes of L. donovani via a resazurin-based assay and compared to image-based microscopy. Using the resazurin-based assay, all evaluated therapies showed reproducible anti-leishmanial activity against the parasite's different life-stages. These results were consistent to the traditional image-based technique. The gold standard of therapy, AMB, showed the highest potency against intracellular L. donovani, and was further evaluated for combinatorial effects with the HDTs. Among the combinations analyzed, pathogen-directed AMB and host-directed AR-12 showed a synergistic reduction of intracellular L. donovani compared to individual treatments. The modified resazurin assay used in this study demonstrated a useful technique to measure new anti-leishmanial drugs against both intracellular and extracellular parasites. The synergistic interactions between pathogen-directed AMB and host-directed AR-12 showed a great promise to combat VL, with the potential to reduce the emergence of drug-resistant strains.
Collapse
Affiliation(s)
- M Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Monica M Johnson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Robert J Tokarski
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Abhay R Satoskar
- Department of Pathology, Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - James R Fuchs
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
23
|
Systematic Review of Host-Mediated Activity of Miltefosine in Leishmaniasis through Immunomodulation. Antimicrob Agents Chemother 2019; 63:AAC.02507-18. [PMID: 31036692 PMCID: PMC6591591 DOI: 10.1128/aac.02507-18] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/22/2019] [Indexed: 12/12/2022] Open
Abstract
Host immune responses are pivotal for the successful treatment of the leishmaniases, a spectrum of infections caused by Leishmania parasites. Previous studies speculated that augmenting cytokines associated with a type 1 T-helper cell (Th1) response is necessary to combat severe forms of leishmaniasis, and it has been hypothesized that the antileishmanial drug miltefosine is capable of immunomodulation and induction of Th1 cytokines. Host immune responses are pivotal for the successful treatment of the leishmaniases, a spectrum of infections caused by Leishmania parasites. Previous studies speculated that augmenting cytokines associated with a type 1 T-helper cell (Th1) response is necessary to combat severe forms of leishmaniasis, and it has been hypothesized that the antileishmanial drug miltefosine is capable of immunomodulation and induction of Th1 cytokines. A better understanding of the immunomodulatory effects of miltefosine is central to providing a rationale regarding synergistic mechanisms of activity to combine miltefosine optimally with other conventional and future antileishmanials that are currently under development. Therefore, a systematic literature search was performed to evaluate to what extent and how miltefosine influences the host Th1 response. Miltefosine’s effects observed in both a preclinical and a clinical context associated with immunomodulation in the treatment of leishmaniasis are evaluated in this review. A total of 27 studies were included in the analysis. Based on the current evidence, miltefosine is not only capable of inducing direct parasite killing but also of modulating the host immunity. Our findings suggest that miltefosine-induced activation of Th1 cytokines, particularly represented by increased gamma interferon (IFN-γ) and interleukin 12 (IL-12), is essential to prevail over the Leishmania-driven Th2 response. Differences in miltefosine-induced host-mediated effects between in vitro, ex vivo, animal model, and human studies are further discussed. All things considered, an effective treatment with miltefosine is acquired by enhanced functional Th1 cytokine responses and may further be enhanced in combination with immunostimulatory agents.
Collapse
|
24
|
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
|
25
|
Heterogeneity of humoral immune response to Leishmania tropica in an experimental model. Parasitol Res 2019; 118:1231-1237. [PMID: 30778754 DOI: 10.1007/s00436-019-06256-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 02/11/2019] [Indexed: 10/27/2022]
Abstract
Humoral (antibody) response is an important part of immunity against pathogens. Despite the clear role of cell-mediated immune response in protection against leishmaniasis, the role of humoral responses is challenging. There is very limited data regarding humoral immune response against Leishmania tropica which is the causative agent of human cutaneous leishmaniasis in many parts of the world. Here, we have compared pathogenicity and antibody response against six Iranian Leishmania tropica isolates in BALB/c mice. A Leishmania major isolate was used for comparison. The parasites were injected into the mice followed by the evaluation of the lesion development, parasite load, and antibody responses (IgG1 and IgG2a). Our findings showed that some isolates caused the large lesions and high parasite load in the spleen and lymph node, while other isolates led to no lesion, no splenic parasitism, and low parasite load in the lymph node. The more pathogenic isolates induced higher antibody responses (IgG1 and IgG2a). Our results indicated that there is substantial heterogeneity among various Leishmania tropica isolates regarding the humoral immune response as well as the pathogenicity.
Collapse
|
26
|
Feasibility Analysis of Interleukin-13 as a Target for a Therapeutic Vaccine. Vaccines (Basel) 2019; 7:vaccines7010020. [PMID: 30759882 PMCID: PMC6466196 DOI: 10.3390/vaccines7010020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The development of therapeutic vaccines requires thorough knowledge of potential hazards associated with long-term inactivation of self-proteins. Among potential targets, interleukin 13 (IL-13) merits consideration, as monoclonal antibodies disrupting IL-13 signaling are proving to be exceedingly effective in common conditions such as atopic dermatitis. OBJECTIVE Given the mass publication of scientific data, an appraisal of safety aspects is challenging. METHODS We here provide a three-fold approach to survey clinically relevant information on off-target effects, both adverse and beneficial, that may potentially be encountered in patients undergoing long-term IL-13 inactivation. First, we review non-clinical data in vivo and in vitro. Second, we summarize safety data accumulating from patients dosed with anti-IL-13 drugs. Third, we exploit human mutation data as well as emerging large-scale genetic datasets (global exome data from 60,000 patients) to obtain information on any association of IL-13-inactivating genetic variants with disease states. In addition, we: (1) dissect the precise efficacy signals obtained with various drugs targeting IL-13 and/or IL-4, and (2) summarize unintended, but potentially beneficial effects of prolonged IL-13 inactivation on several functional systems. RESULTS Prolonged repression of IL-13 in several thousand patients so far has not uncovered any non-redundant functions of IL-13 in immune defense. Furthermore, missense mutations in the key genes IL-13, IL-13Rα1, IL-13Rα2, IL-4, IL-4Rα are common, while no case reports have been published on any immune deficiency or increased risk of neoplastic disease associated with such mutations, suggesting that these genes do not harbor non-redundant roles in adult outbred humans. In terms of efficacy, data from clinically used drugs strongly suggest that targeting IL-13 only, as opposed to IL-13 and IL-4, may be effective in eczema while being more selective. Importantly, several lines of evidence suggest that inhibition of IL-13 may in fact harbor potentially beneficial effects on non-targeted systems, including glucose metabolism, hepatic fibrosis, and atherosclerosis, suggesting that respective outcomes should be systematically captured in patients dosed with IL-13 interfering drugs. Collectively, available evidence suggests that IL-13 may fulfill safety requirements required for the target of a therapeutic vaccine.
Collapse
|
27
|
Astawa INM, Oka IBM, Dwinata IM. Antibody immunoglobulin G1 and immunoglobulin G2a responses against some cystic fluid proteins of Cysticercus bovis in Balb/c mice. Vet World 2018; 11:1641-1647. [PMID: 30587902 PMCID: PMC6303487 DOI: 10.14202/vetworld.2018.1641-1647] [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: 07/17/2018] [Accepted: 10/11/2018] [Indexed: 01/23/2023] Open
Abstract
Background and Aim: Immunoglobulin (Ig) G1 and IgG2a are the surrogate markers respectively for humoral and cellular immune responses of hosts against antigens including cystic fluid proteins of Cysticercus bovis. A study was conducted to investigate the IgG1 and IgG2a responses of Balb/c mice against some individual cystic fluid proteins of C. bovis in an effort to determine the roles of each protein in inducing the humoral and cellular immune responses in host. Materials and Methods: Individual p71, p31, and p14 proteins of C. bovis were purified by separation of the proteins using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and elution of individual proteins from the gel. Six female Balb/c mice were immunized 4 times at 10-day intervals with the crude cystic fluid proteins, and sera were collected for the measurement of IgG1 and IgG2a levels against the individual proteins. Sera samples collected before the first immunization were used as negative antibody control, sera samples collected after the fourth immunization were used as positive antibody control, and crude cystic fluid protein was used as positive antigen control. Results: All immunized mice were immune to p71, p31, p14, and crude cystic fluid proteins of C. bovis. The crude cystic fluid proteins of C. bovis induced a higher IgG2a than IgG1 level following the first and the second immunizations but switched into a higher IgG1 than IgG2a level following the fourth immunization. Protein 71 kDa (p71) induced a higher IgG2a than IgG1 level following the fourth immunization. In contrast, p14 induced a higher IgG1 than IgG2a level following the fourth immunization. Low and balance IgG1 and IgG2a levels against p31 were observed following the first to the fourth immunizations. Conclusion: Using IgG1 and IgG2a levels as the surrogate markers, it appears that cystic fluid antigens of C. bovis induce both humoral and cellular immune responses in Balb/c mice. The p71 appears to be a better inducer of cellular immune response, whereas p14 is a better inducer of humoral immune response of mice.
Collapse
Affiliation(s)
- I Nyoman Mantik Astawa
- Laboratory of Immunology, Faculty of Veterinary Medicine, Udayana University, Denpasar Bali 80232, Indonesia
| | - Ida Bagus Made Oka
- Laboratory of Parasitology, Faculty of Veterinary Medicine, Udayana University, Bali 80232, Indonesia
| | - I Made Dwinata
- Laboratory of Parasitology, Faculty of Veterinary Medicine, Udayana University, Bali 80232, Indonesia
| |
Collapse
|
28
|
Efficacy of Four Solanum spp. Extracts in an Animal Model of Cutaneous Leishmaniasis. MEDICINES 2018; 5:medicines5020049. [PMID: 29874837 PMCID: PMC6023388 DOI: 10.3390/medicines5020049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/15/2018] [Accepted: 05/25/2018] [Indexed: 11/16/2022]
Abstract
Background: Leishmaniasis is a complex protozoa disease caused by Leishmania genus (Trypanosomatidae family). Currently, there have been renewed interests worldwide in plants as pharmaceutical agents. In this study, the in vivo efficacy of Solanum spp. is assessed in an L. amazonensis BALB/c mice model for experimental cutaneous leishmaniasis. Methods: Animals were infected with 5 × 10⁶ metacyclic promastigotes and 30-day post-infection, a treatment with 30 mg/kg of Solanum extracts or Glucantime® (GTM) was applied intralesionally every four days to complete 5 doses. Results: Neither death nor loss of weight higher than 10% was observed. All the tested extracts were able to control the infection, compared with the infected and untreated group. Solanum havanense Jacq. extract showed the highest efficacy and was superior (p < 0.05) to GTM. Solanum myriacanthum Dunal., S. nudum Dunal. and S. seaforthianum Andr. extracts demonstrated a similar effect (p > 0.05) to GTM. An increase of IFN-γ (p < 0.05) was displayed only by animals treated with S. nudum compared to the group treated with a vehicle, while no differences (p > 0.05) were observed for IL-12. Conclusions:In vivo effects of Solanum extracts were demonstrated, suggesting that this genus could be further explored as a new antileishmanial alternative.
Collapse
|
29
|
Grayfer L, Kerimoglu B, Yaparla A, Hodgkinson JW, Xie J, Belosevic M. Mechanisms of Fish Macrophage Antimicrobial Immunity. Front Immunol 2018; 9:1105. [PMID: 29892285 PMCID: PMC5985312 DOI: 10.3389/fimmu.2018.01105] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/02/2018] [Indexed: 12/13/2022] Open
Abstract
Overcrowding conditions and temperatures shifts regularly manifest in large-scale infections of farmed fish, resulting in economic losses for the global aquaculture industries. Increased understanding of the functional mechanisms of fish antimicrobial host defenses is an important step forward in prevention of pathogen-induced morbidity and mortality in aquaculture setting. Like other vertebrates, macrophage-lineage cells are integral to fish immune responses and for this reason, much of the recent fish immunology research has focused on fish macrophage biology. These studies have revealed notable similarities as well as striking differences in the molecular strategies by which fish and higher vertebrates control their respective macrophage polarization and functionality. In this review, we address the current understanding of the biological mechanisms of teleost macrophage functional heterogeneity and immunity, focusing on the key cytokine regulators that control fish macrophage development and their antimicrobial armamentarium.
Collapse
Affiliation(s)
- Leon Grayfer
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | - Baris Kerimoglu
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | - Amulya Yaparla
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | | | - Jiasong Xie
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Miodrag Belosevic
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
30
|
Almeida APMM, Machado LFM, Doro D, Nascimento FC, Damasceno L, Gazzinelli RT, Fernandes AP, Junqueira C. New Vaccine Formulations Containing a Modified Version of the Amastigote 2 Antigen and the Non-Virulent Trypanosoma cruzi CL-14 Strain Are Highly Antigenic and Protective against Leishmania infantum Challenge. Front Immunol 2018; 9:465. [PMID: 29599776 PMCID: PMC5863692 DOI: 10.3389/fimmu.2018.00465] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/21/2018] [Indexed: 12/21/2022] Open
Abstract
Visceral leishmaniasis (VL) is a major public health issue reported as the second illness in mortality among all tropical diseases. Clinical trials have shown that protection against VL is associated with robust T cell responses, especially those producing IFN-γ. The Leishmania amastigote 2 (A2) protein has been repeatedly described as immunogenic and protective against VL in different animal models; it is recognized by human T cells, and it is also commercially available in a vaccine formulation containing saponin against canine VL. Moving toward a more appropriate formulation for human vaccination, here, we tested a new optimized version of the recombinant protein (rA2), designed for Escherichia coli expression, in combination with adjuvants that have been approved for human use. Moreover, aiming at improving the cellular immune response triggered by rA2, we generated a recombinant live vaccine vector using Trypanosoma cruzi CL-14 non-virulent strain, named CL-14 A2. Mice immunized with respective rA2, adsorbed in Alum/CpG B297, a TLR9 agonist recognized by mice and human homologs, or with the recombinant CL-14 A2 parasites through homologous prime-boost protocol, were evaluated for antigen-specific immune responses and protection against Leishmania infantum promastigote challenge. Immunization with the new rA2/Alum/CpG formulations and CL-14 A2 transgenic vectors elicited stronger cellular immune responses than control groups, as shown by increased levels of IFN-γ, conferring protection against L. infantum challenge. Interestingly, the use of the wild-type CL-14 alone was enough to boost immunity and confer protection, confirming the previously reported immunogenic potential of this strain. Together, these results support the success of both the newly designed rA2 antigen and the ability of T. cruzi CL-14 to induce strong T cell-mediated immune responses against VL in animal models when used as a live vaccine vector. In conclusion, the vaccination strategies explored here reveal promising alternatives for the development of new rA2 vaccine formulations to be translated human clinical trials.
Collapse
Affiliation(s)
- Ana Paula M M Almeida
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leopoldo F M Machado
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Daniel Doro
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Frederico C Nascimento
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Ricardo Tostes Gazzinelli
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.,Division of Infectious Disease, University of Massachusetts Medical School, Worcester, MA, United States
| | - Ana Paula Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
31
|
Schleicher U, Liese J, Justies N, Mischke T, Haeberlein S, Sebald H, Kalinke U, Weiss S, Bogdan C. Type I Interferon Signaling Is Required for CpG-Oligodesoxynucleotide-Induced Control of Leishmania major, but Not for Spontaneous Cure of Subcutaneous Primary or Secondary L. major Infection. Front Immunol 2018; 9:79. [PMID: 29459858 PMCID: PMC5807663 DOI: 10.3389/fimmu.2018.00079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/11/2018] [Indexed: 01/11/2023] Open
Abstract
We previously showed that in mice infected with Leishmania major type I interferons (IFNs) initiate the innate immune response to the parasite at day 1 and 2 of infection. Here, we investigated which type I IFN subtypes are expressed during the first 8 weeks of L. major infection and whether type I IFNs are essential for a protective immune response and clinical cure of the disease. In self-healing C57BL/6 mice infected with a high dose of L. major, IFN-α4, IFN-α5, IFN-α11, IFN-α13, and IFN-β mRNA were most prominently regulated during the course of infection. In C57BL/6 mice deficient for IFN-β or the IFN-α/β-receptor chain 1 (IFNAR1), development of skin lesions and parasite loads in skin, draining lymph node, and spleen was indistinguishable from wild-type (WT) mice. In line with the clinical findings, C57BL/6 IFN-β−/−, IFNAR1−/−, and WT mice exhibited similar mRNA expression levels of IFN-γ, interleukin (IL)-4, IL-12, IL-13, inducible nitric oxide synthase, and arginase 1 during the acute and late phase of the infection. Also, myeloid dendritic cells from WT and IFNAR1−/− mice produced comparable amounts of IL-12p40/p70 protein upon exposure to L. major in vitro. In non-healing BALB/c WT mice, the mRNAs of IFN-α subtypes (α2, α4, α5, α6, and α9) were rapidly induced after high-dose L. major infection. However, genetic deletion of IFNAR1 or IFN-β did not alter the progressive course of infection seen in WT BALB/c mice. Finally, we tested whether type I IFNs and/or IL-12 are required for the prophylactic effect of CpG-oligodesoxynucleotides (ODN) in BALB/c mice. Local and systemic administration of CpG-ODN 1668 protected WT and IFN-β−/− mice equally well from progressive leishmaniasis. By contrast, the protective effect of CpG-ODN 1668 was lost in BALB/c IFNAR1−/− (despite a sustained suppression of IL-4) and in BALB/c IL-12p35−/− mice. From these data, we conclude that IFN-β and IFNAR1 signaling are dispensable for a curative immune response to L. major in C57BL/6 mice and irrelevant for disease development in BALB/c mice, whereas IL-12 and IFN-α subtypes are essential for the disease prevention by CpG-ODNs in this mouse strain.
Collapse
Affiliation(s)
- Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Jan Liese
- Abteilung Mikrobiologie und Hygiene, Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Nicole Justies
- Abteilung Mikrobiologie und Hygiene, Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Thomas Mischke
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simone Haeberlein
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Heidi Sebald
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ulrich Kalinke
- Institut für Experimentelle Infektionsforschung, TWINCORE, Zentrum für Experimentelle und Klinische Infektionsforschung, eine Gemeinschaftseinrichtung vom Helmholtz Zentrum für Infektionsforschung und der Medizinischen Hochschule Hannover, Hannover, Germany
| | - Siegfried Weiss
- Abteilung für Molekulare Immunologie, Helmholtz Zentrum für Infektionsforschung, Braunschweig, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
32
|
Reduced pathogenicity of fructose-1,6-bisphosphatase deficient Leishmania donovani and its use as an attenuated strain to induce protective immunogenicity. Vaccine 2018; 36:1190-1202. [PMID: 29395522 DOI: 10.1016/j.vaccine.2018.01.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 11/25/2017] [Accepted: 01/11/2018] [Indexed: 01/08/2023]
Abstract
Currently, there is no approved vaccine for visceral leishmaniasis (VL) caused by L. donovani. The ability to manipulate Leishmania genome by eliminating or introducing genes necessary for parasites' survival considered as the powerful strategy to generate the live attenuated vaccine. In the present study fructose-1,6-bisphosphatase (LdFBPase) gene deleted L. donovani (Δfbpase) was generated using homologous gene replacement strategy. Though LdFBPase gene deletion (Δfbpase) does not affect the growth of parasite in the promastigote form but axenic amastigotes display a marked reduction in their capacity to multiply in vitro inside macrophages and in vivo in Balb/c mice. Though Δfbpase L. donovani parasite persisted in BALB/c mice up to 12 weeks but was unable to cause infection, we tested its ability to protect against a virulent L. donovani challenge. Notably, intraperitoneal immunisation with live Δfbpase parasites displayed the reduction of parasites load in mice spleen and liver post challenge. Moreover, immunised BALB/c mice showed a reversal of T cell anergy and high levels of NO production that result in the killing of the parasite. A significant, correlation was found between parasite clearance and elevated IFNγ, IL12, and IFNγ/IL10 ratio compared to IL10 and TGFβ in immunised and challenged mice. Results suggested the generation of protective Th1 type immune response which induced significant parasite clearance at 12-week, as well as 16 weeks post, challenged immunised mice, signifying sustained immunity. Therefore, we propose that Δfbpase L. donovani parasites can be a live attenuated vaccine candidate for VL and a good model to understand the correlatives of protection in visceral leishmaniasis.
Collapse
|
33
|
El‐Bendary M, Neamatallah M, Elalfy H, Besheer T, El‐Setouhy M, Kasim N, Abou El‐Khier NT, Kamel E, Eladl A, El‐Waseef A, Abdel‐Aziz AF, Esmat G. Association of interferon gamma gene polymorphism and susceptibility to hepatitis C virus infection in Egyptian patients: A multicenter, family-based study. JGH Open 2017; 1:140-147. [PMID: 30483551 PMCID: PMC6207041 DOI: 10.1002/jgh3.12024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/17/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIM Polymorphisms in some genes may influence the persistence of hepatitis C virus (HCV) infection, clinical outcome, HCV replication, and liver damage. This study was conducted to investigate the role of the interferon gamma (IFN-γ) gene at (+874 T/A, -764 G/C, -179 C/A) single-nucleotide polymorphisms (SNPs) and its receptor (IFN-γR2) at (rs 2786067 A/C) SNP in the susceptibility of Egyptian families to HCV infection with high-resolution techniques. METHODS In total, 517 Egyptian families, with 2246 subjects, were recruited to this study from the Upper and Lower Egypt governorates and were classified into three groups: 1034 patients with chronic hepatitis C virus, 108 subjects with spontaneous virus clearance (SVC), and 1104 subjects as a healthy control group. All subjects were genotyped for (+874 T/A, rs2430561, -764 G/C, rs2069707, -179 C/A, rs2069709, and rs 27860067, A/C) SNPs of the IFN-γ gene using the allelic discrimination real-time polymerase chain reaction technique and were confirmed using sequence-based typing. RESULTS The carriage of T allele of (+874) IFN-γ is a risky allele and was significantly higher in chronic hepatitis C more than other two groups (odds ratio [OR]: 2.6646, P < 0.0002). On the other hand, the C allele of (-764, rs2069707) is a protective allele and was higher in SVC than the other two groups (OR: 0.2709, P < 0.0001). However, both (-179 C/A, rs 2069709) and (rs 27860067, A/C) SNPs are not polymorphic enough to be studied in the Egyptian population. CONCLUSIONS HCV infection is associated with the T allele of (+874 rs2430561), while SVC of HCV is associated with the C allele of (-764, rs2069707) of the IFN-γ gene.
Collapse
Affiliation(s)
- Mahmoud El‐Bendary
- Tropical Medicine and Hepatology Department, Mansoura Faculty of MedicineMansoura UniversityMansouraEgypt
| | - Mustafa Neamatallah
- Medical Biochemistry Department, Mansoura Faculty of MedicineMansoura UniversityMansouraEgypt
| | - Hatem Elalfy
- Tropical Medicine and Hepatology Department, Mansoura Faculty of MedicineMansoura UniversityMansouraEgypt
| | - Tarek Besheer
- Tropical Medicine and Hepatology Department, Mansoura Faculty of MedicineMansoura UniversityMansouraEgypt
| | - Maged El‐Setouhy
- Department of Community, Environmental and Occupational Medicine, Ain‐Shams Faculty of MedicineAin‐Shams UniversityCairoEgypt
- Substance Abuse Research Center (SARC)Jazan UniversityJazanKingdom of Saudi Arabia
| | - Nihal Kasim
- Biochemistry Department, Faculty of ScienceMansoura UniversityMansouraEgypt
| | - Noha T Abou El‐Khier
- Medical Microbiology and Immunology Department, Faculty of MedicineMansoura UniversityMansouraEgypt
| | - Emily Kamel
- Public Health and Preventive Medicine Department, Mansoura Faculty of MedicineMansoura UniversityMansouraEgypt
| | - Abdel‐Hamid Eladl
- Internal Medicine Department, Alazhar Faculty of MedicineAssiut UniversityAssiutEgypt
| | - Ahmad El‐Waseef
- Biochemistry Department, Faculty of ScienceMansoura UniversityMansouraEgypt
| | | | - Gamal Esmat
- Tropical Medicine and Hepatology Department, Cairo Faculty of MedicineCairo UniversityGizaEgypt
| |
Collapse
|
34
|
Das A, Ranganathan V, Umar D, Thukral S, George A, Rath S, Bal V. Effector/memory CD4 T cells making either Th1 or Th2 cytokines commonly co-express T-bet and GATA-3. PLoS One 2017; 12:e0185932. [PMID: 29088218 PMCID: PMC5663332 DOI: 10.1371/journal.pone.0185932] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 09/21/2017] [Indexed: 11/23/2022] Open
Abstract
Naïve CD4 T (NCD4T) cells post-activation undergo programming for inducible production of cytokines leading to generation of memory cells with various functions. Based on cytokine based polarization of NCD4T cells in vitro, programming for either ‘Th1’ (interferon-gamma [IFNg]) or ‘Th2’ (interleukin [IL]-4/5/13) cytokines is thought to occur via mutually exclusive expression and functioning of T-bet or GATA-3 transcription factors (TFs). However, we show that a high proportion of mouse and human memory-phenotype CD4 T (MCD4T) cells generated in vivo which expressed either Th1 or Th2 cytokines commonly co-expressed T-bet and GATA-3. While T-bet levels did not differ between IFNg-expressing and IL-4/5/13-expressing MCD4T cells, GATA-3 levels were higher in the latter. These observations were also confirmed in MCD4T cells from FVB/NJ or aged C57BL/6 or IFNg-deficient mice. While MCD4T cells from these strains showed greater Th2 commitment than those from young C57BL/6 mice, pattern of co-expression of TF was similar. Effector T cells generated in vivo following immunization also showed TF co-expression in Th1 or Th2 cytokine producing cells. We speculated that the difference in TF expression pattern of MCD4T cells generated in vivo and those generated in cytokine polarized cultures in vitro could be due to relative absence of polarizing conditions during activation in vivo. We tested this by NCD4T cell activation in non-polarizing conditions in vitro. Anti-CD3 and anti-CD28-mediated priming of polyclonal NCD4T cells in vitro without polarizing milieu generated cells that expressed either IFNg or IL-4/5/13 but not both, yet both IFNg- and IL-4/5/13-expressing cells showed upregulation of both TFs. We also tested monoclonal T cell populations activated in non-polarizing conditions. TCR-transgenic NCD4T cells primed in vitro by cognate peptide in non-polarizing conditions which expressed either IFNg or IL-4/5/13 also showed a high proportion of cells co-expressing TFs, and their cytokine commitment varied depending on genetic background or priming conditions, without altering pattern of TF co-expression. Thus, the model of mutually antagonistic differentiation programs driven by mutually exclusively expressed T-bet or GATA-3 does not completely explain natural CD4 T cell priming outcomes.
Collapse
Affiliation(s)
| | | | - Danish Umar
- National Institute of Immunology, New Delhi, India
| | | | - Anna George
- National Institute of Immunology, New Delhi, India
| | | | - Vineeta Bal
- National Institute of Immunology, New Delhi, India
- * E-mail:
| |
Collapse
|
35
|
The Th1/Th17 balance dictates the fibrosis response in murine radiation-induced lung disease. Sci Rep 2017; 7:11586. [PMID: 28912510 PMCID: PMC5599556 DOI: 10.1038/s41598-017-11656-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/25/2017] [Indexed: 01/02/2023] Open
Abstract
Radiotherapy can result in lung diseases pneumonitis or fibrosis dependent on patient susceptibility. Herein we used inbred and genetically altered mice to investigate whether the tissue adaptive immune response to radiation injury influences the development of radiation-induced lung disease. Six inbred mouse strains were exposed to 18 Gy whole thorax irradiation and upon respiratory distress strains prone to pneumonitis with fibrosis presented an increased pulmonary frequency of Thelper (Th)17 cells which was not evident in strains prone solely to pneumonitis. The contribution of Th17 cells to fibrosis development was supported as the known enhanced fibrosis of toll-like receptor 2&4 deficient mice, compared to C57BL/6J mice, occurred with earlier onset neutrophilia, and with increased levels of pulmonary Th17, but not Th1, cells following irradiation. Irradiated Il17−/− mice lacked Th17 cells, and were spared both fibrosis and pneumonitis, as they survived to the end of the experiment with a significantly increased pulmonary Th1 cell frequency, only. Interferon-γ−/− mice, deficient in Th1 cells, developed a significantly enhanced fibrosis response compared to that of C57BL/6J mice. The tissue adaptive immune response influences the pulmonary disease response to radiotherapy, as an increased Th17 cell frequency enhanced and a Th1 response spared, fibrosis in mice.
Collapse
|
36
|
Ahn JJ, Selvanantham T, Zhang MA, Mallevaey T, Dunn SE. Experimental Infection with Listeria monocytogenes as a Model for Studying Host Interferon-γ Responses. J Vis Exp 2016. [PMID: 27911410 DOI: 10.3791/54554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
L. monocytogenes is a gram-positive bacterium that is a cause of food borne disease in humans. Experimental infection of mice with this pathogen has been highly informative on the role of innate and adaptive immune cells and specific cytokines in host immunity against intracellular pathogens. Production of IFN-γ by innate cells during sublethal infection with L. monocytogenes is important for activating macrophages and early control of the pathogen1-3. In addition, IFN-γ production by adaptive memory lymphocytes is important for priming the activation of innate cells upon reinfection4. The L. monocytogenes infection model thus serves as a great tool for investigating whether new therapies that are designed to increase IFN-γ production have an impact on IFN-γ responses in vivo and have productive biological effects such as increasing bacterial clearance or improving mouse survival from infection. Described here is a basic protocol for how to conduct intraperitoneal infections of C57BL/6J mice with the EGD strain of L. monocytogenes and to measure IFN-γ production by NK cells, NKT cells, and adaptive lymphocytes by flow cytometry. In addition, procedures are described to: (1) grow and prepare the bacteria for inoculation, (2) measure bacterial load in the spleen and liver, and (3) measure animal survival to endpoints. Representative data are also provided to illustrate how this infection model can be used to test the effect of specific agents on IFN-γ responses to L. monocytogenes and survival of mice from this infection.
Collapse
Affiliation(s)
| | | | | | | | - Shannon E Dunn
- Department of Immunology, University of Toronto; Toronto General Research Institute, University Health Network; Women's College Research Institute;
| |
Collapse
|
37
|
Schmidt S, Ullrich E, Bochennek K, Zimmermann SY, Lehrnbecher T. Role of natural killer cells in antibacterial immunity. Expert Rev Hematol 2016; 9:1119-1127. [DOI: 10.1080/17474086.2016.1254546] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
38
|
Schatz V, Strüssmann Y, Mahnke A, Schley G, Waldner M, Ritter U, Wild J, Willam C, Dehne N, Brüne B, McNiff JM, Colegio OR, Bogdan C, Jantsch J. Myeloid Cell-Derived HIF-1α Promotes Control of Leishmania major. THE JOURNAL OF IMMUNOLOGY 2016; 197:4034-4041. [PMID: 27798163 DOI: 10.4049/jimmunol.1601080] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/15/2016] [Indexed: 12/30/2022]
Abstract
Hypoxia-inducible factor-1α (HIF-1α), which accumulates in mammalian host organisms during infection, supports the defense against microbial pathogens. However, whether and to what extent HIF-1α expressed by myeloid cells contributes to the innate immune response against Leishmania major parasites is unknown. We observed that Leishmania-infected humans and L. major-infected C57BL/6 mice exhibited substantial amounts of HIF-1α in acute cutaneous lesions. In vitro, HIF-1α was required for leishmanicidal activity and high-level NO production by IFN-γ/LPS-activated macrophages. Mice deficient for HIF-1α in their myeloid cell compartment had a more severe clinical course of infection and increased parasite burden in the skin lesions compared with wild-type controls. These findings were paralleled by reduced expression of type 2 NO synthase by lesional CD11b+ cells. Together, these data illustrate that HIF-1α is required for optimal innate leishmanicidal immune responses and, thereby, contributes to the cure of cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Valentin Schatz
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Yannic Strüssmann
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Alexander Mahnke
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie, und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Gunnar Schley
- Medizinische Klinik 4, Nephrologie und Hypertensiologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Maximilian Waldner
- Medizinische Klinik 1, Gastroenterologie, Pneumologie und Endokrinologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Uwe Ritter
- Institute of Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Jens Wild
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Carsten Willam
- Medizinische Klinik 4, Nephrologie und Hypertensiologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Nathalie Dehne
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany; and
| | - Bernhard Brüne
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany; and
| | - Jennifer M McNiff
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06510
| | - Oscar R Colegio
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06510
| | - Christian Bogdan
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie, und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany;
| |
Collapse
|
39
|
Immunization with KMP11-NTGP96-GFP Fusion of Leishmania major Induced Th1 Platform Immune Response in Susceptible BALB/c mice. Jundishapur J Microbiol 2016. [DOI: 10.5812/jjm.36186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
40
|
Miahipour A, Haji-Fatahaliha M, Keshavarz H, Gharavi MJ, Mohamadi H, Babaloo Z, Rafati S, Younesi V, Hosseini M, Yousefi M. T Helper 1 (Th1), Th2, and Th17 Responses to Leishmania major Lipophosphoglycan 3. Immunol Invest 2016; 45:692-702. [PMID: 27611455 DOI: 10.1080/08820139.2016.1208217] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leishmania major is the main causal agent of cutaneous leishmaniasis (CL) that remains a serious public health concern in many tropical and subtropical countries. A long-lasting protective vaccine against leishmaniasis remains as a medical unmet need. Lipophosphoglycan 3 (LPG3) is one of the class II LPG genes from HSP90 family involved in the host immune responses. The aim of the present study is to investigate the capability of recombinant LPG3 (rLPG3) to induce Th1, Th2, Th17 responses. The results showed that rLPG3 in moderate and high concentrations significantly induced expression of Th1 lineage-specific transcription factor (T-bet) and cytokine (IFN-γ)(P < 0.05). Moreover, the Th1-stimulating effect of rLPG3 was confirmed by significant induction of IFN-γ secretion from treated T cells (P < 0.01). However, no significant effect of rLPG3 on Th2 and Th17 lineage cells was observed even in high concentration. Our findings demonstrate that rLPG3 induces Th1, but not Th2 and Th17, lineage responses. Further studies are needed to investigate adjuvant properties of rLPG3 for leishmania therapy.
Collapse
Affiliation(s)
- Abolfazl Miahipour
- a Department of Medical Parasitology and Mycology , School of Medicine, Alborz University of Medical Sciences , Karaj , Iran
| | - Mostafa Haji-Fatahaliha
- b Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- c Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- d Department of Immunology, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Hossein Keshavarz
- e Department of Medical Parasitology and Mycology, School of Public Health , Tehran University of Medical Sciences , Tehran , Iran
- f Center for Research of Endemic Parasites of Iran (CREPI) , Tehran University of Medical Sciences , Tehran , Iran
| | - Mohammad Javad Gharavi
- a Department of Medical Parasitology and Mycology , School of Medicine, Alborz University of Medical Sciences , Karaj , Iran
| | - Hamed Mohamadi
- b Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- c Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- d Department of Immunology, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Zohre Babaloo
- b Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- c Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- d Department of Immunology, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Sima Rafati
- g Molecular Immunology and Vaccine Research Lab , Pasteur Institute of Iran , Tehran , Iran
| | - Vahid Younesi
- a Department of Medical Parasitology and Mycology , School of Medicine, Alborz University of Medical Sciences , Karaj , Iran
| | - Maryam Hosseini
- b Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- c Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- d Department of Immunology, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mehdi Yousefi
- b Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- c Immunology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
- d Department of Immunology, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
41
|
Lopes MEM, Carneiro MBH, dos Santos LM, Vieira LQ. Indigenous microbiota and Leishmaniasis. Parasite Immunol 2015; 38:37-44. [DOI: 10.1111/pim.12279] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/17/2015] [Indexed: 12/14/2022]
Affiliation(s)
- M. E. M. Lopes
- Departamento de Bioquímica e Imunologia; ICB; Universidade Federal de Minas Gerais; Belo Horizonte MG Brazil
| | - M. B. H. Carneiro
- Departamento de Bioquímica e Imunologia; ICB; Universidade Federal de Minas Gerais; Belo Horizonte MG Brazil
| | - L. M. dos Santos
- Departamento de Bioquímica e Imunologia; ICB; Universidade Federal de Minas Gerais; Belo Horizonte MG Brazil
| | - L. Q. Vieira
- Departamento de Bioquímica e Imunologia; ICB; Universidade Federal de Minas Gerais; Belo Horizonte MG Brazil
| |
Collapse
|
42
|
Carneiro MBH, Lopes MEDM, Vaz LG, Sousa LMA, dos Santos LM, de Souza CC, Campos ACDA, Gomes DA, Gonçalves R, Tafuri WL, Vieira LQ. IFN-γ-Dependent Recruitment of CD4(+) T Cells and Macrophages Contributes to Pathogenesis During Leishmania amazonensis Infection. J Interferon Cytokine Res 2015; 35:935-47. [PMID: 26401717 PMCID: PMC4683564 DOI: 10.1089/jir.2015.0043] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/09/2015] [Indexed: 12/21/2022] Open
Abstract
Interferon gamma (IFN-γ) is a key factor in the protection of hosts against intracellular parasites. This cytokine induces parasite killing through nitric oxide and reactive oxygen species production by phagocytes. Surprisingly, during Leishmania amazonensis infection, IFN-γ plays controversial roles. During in vitro infections, IFN-γ induces the proliferation of the amastigote forms of L. amazonensis. However, this cytokine is not essential at the beginning of an in vivo infection. It is not clear why IFN-γ does not mediate protection during the early stages of infection. Thus, the aim of our study was to investigate the role of IFN-γ during L. amazonensis infection. We infected IFN-γ(-/-) mice in the footpad and followed the development of leishmaniasis in these mice compared with that in WT mice. CD4(+) T lymphocytes and macrophages migrated earlier to the site of infection in the WT mice, and the earlier migration of these 2 cell types was associated with lesion development and parasite growth, respectively. These differences in the infiltrate populations were explained by the increased expression of chemokines in the lesions of the WT mice. Thus, we propose that IFN-γ plays a dual role during L. amazonensis infection; it is an important inducer of effector mechanisms, particularly through inducible nitric oxide synthase expression, and conversely, it is a mediator of inflammation and pathogenesis through the induction of the expression of chemokines. Our data provided evidence for a pathogenic effect of IFN-γ production during leishmaniasis that was previously unknown.
Collapse
Affiliation(s)
- Matheus Batista Heitor Carneiro
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mateus Eustáquio de Moura Lopes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo Gomes Vaz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Louisa Maria Andrade Sousa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Liliane Martins dos Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carolina Carvalho de Souza
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Carolina de Angelis Campos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Dawidson Assis Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Gonçalves
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Wagner Luiz Tafuri
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leda Quercia Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
43
|
Gupta G, Peine KJ, Abdelhamid D, Snider H, Shelton AB, Rao L, Kotha SR, Huntsman AC, Varikuti S, Oghumu S, Naman CB, Pan L, Parinandi NL, Papenfuss TL, Kinghorn AD, Bachelder EM, Ainslie KM, Fuchs JR, Satoskar AR. A Novel Sterol Isolated from a Plant Used by Mayan Traditional Healers Is Effective in Treatment of Visceral Leishmaniasis Caused by Leishmania donovani. ACS Infect Dis 2015; 1:497-506. [PMID: 27623316 DOI: 10.1021/acsinfecdis.5b00081] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Visceral leishmaniasis (VL), caused by the protozoan parasite Leishmania donovani, is a global health problem affecting millions of people worldwide. Treatment of VL largely depends on therapeutic drugs such as pentavalent antimonials, amphotericin B, and others, which have major drawbacks due to drug resistance, toxicity, and high cost. In this study, for the first time, we have successfully demonstrated the synthesis and antileishmanial activity of the novel sterol pentalinonsterol (PEN), which occurs naturally in the root of a Mexican medicinal plant, Pentalinon andrieuxii. In the experimental BALB/c mouse model of VL induced by infection with L. donovani, intravenous treatment with liposome-encapsulated PEN (2.5 mg/kg) led to a significant reduction in parasite burden in the liver and spleen. Furthermore, infected mice treated with liposomal PEN showed a strong host-protective TH1 immune response characterized by IFN-γ production and formation of matured hepatic granulomas. These results indicate that PEN could be developed as a novel drug against VL.
Collapse
Affiliation(s)
- Gaurav Gupta
- Department of Pathology,
The Wexner Medical Center, The Ohio State University, 320 West
10th Avenue, Columbus, Ohio 43210, United States
- Department of Biochemistry and Immunology, School of Medicine of
Ribeirão Preto, University of Sao Paulo, Av. Bandeirantes
3900, 14049-900 Ribeirão Preto, Brazil
| | - Kevin J. Peine
- Molecular,
Cellular and Developmental Biology Graduate Program, The Ohio State University, 484 West 12th Avenue, Columbus, Ohio 43210, United States
- Division of Molecular Pharmaceutics, Eshelman School
of Pharmacy, University of North Carolina, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Dalia Abdelhamid
- Division of Medicinal Chemistry and Pharmacognosy, College
of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
- Department
of Medicinal Chemistry, Minia University, Minia, Egypt
| | - Heidi Snider
- Department of Pathology,
The Wexner Medical Center, The Ohio State University, 320 West
10th Avenue, Columbus, Ohio 43210, United States
| | - Andrew B. Shelton
- Division
of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department
of Internal Medicine, The Wexner Medical Center, The Ohio State University, 473 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Latha Rao
- Division
of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department
of Internal Medicine, The Wexner Medical Center, The Ohio State University, 473 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Sainath R. Kotha
- Division
of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department
of Internal Medicine, The Wexner Medical Center, The Ohio State University, 473 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Andrew C. Huntsman
- Division of Medicinal Chemistry and Pharmacognosy, College
of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Sanjay Varikuti
- Department of Pathology,
The Wexner Medical Center, The Ohio State University, 320 West
10th Avenue, Columbus, Ohio 43210, United States
| | - Steve Oghumu
- Department of Pathology,
The Wexner Medical Center, The Ohio State University, 320 West
10th Avenue, Columbus, Ohio 43210, United States
| | - C. Benjamin Naman
- Division of Medicinal Chemistry and Pharmacognosy, College
of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Li Pan
- Division of Medicinal Chemistry and Pharmacognosy, College
of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Narasimham L. Parinandi
- Division
of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department
of Internal Medicine, The Wexner Medical Center, The Ohio State University, 473 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Tracy L. Papenfuss
- Department of Veterinary Biosciences, College of Veterinary
Medicine, The Ohio State University, 1900 Coffey Road, Columbus, Ohio 43210, United States
- Department
of Pathology, WIL Research, Ashland, Ohio 55805, United States
| | - A. Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College
of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Eric M. Bachelder
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
- Division of Molecular Pharmaceutics, Eshelman School
of Pharmacy, University of North Carolina, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Kristy M. Ainslie
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
- Division of Molecular Pharmaceutics, Eshelman School
of Pharmacy, University of North Carolina, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - James R. Fuchs
- Division of Medicinal Chemistry and Pharmacognosy, College
of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Abhay R. Satoskar
- Department of Pathology,
The Wexner Medical Center, The Ohio State University, 320 West
10th Avenue, Columbus, Ohio 43210, United States
| |
Collapse
|
44
|
Al-Qadhi BN, Musa IS, Al-Mulla Hummadi YMK. Comparative immune study on cutaneous leishmaniasis patients with single and multiple sores. J Parasit Dis 2015; 39:361-70. [PMID: 26345036 PMCID: PMC4554559 DOI: 10.1007/s12639-013-0368-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 10/18/2013] [Indexed: 11/26/2022] Open
Abstract
Ninety-five Iraqi patients with cutaneous leishmaniasis (CL) caused by Leishmania tropica at AL-Karama Hospital in Baghdad were included in this study. Sixty patients were with single sore and the remaining with multiple sores. The study also included 10 atopic patients and 30 healthy individuals as a control group. Cellular and humoral immune response at different stages of the disease activity (early and late) were evaluated by estimation of serum IFN-γ, IL-4 and total IgE antibodies using ELISA kits while, the detection of specific anti leishmanial IgE antibodies was done manually. Specific IgE antibodies were only detected in early CL (<2 months) patients 68 (71.57 %) while, were not detected in late CL, atopic and healthy controls 30 (100 %). The results also showed a positive relationship between this antibody and the number of sores. Th-2 predominates during the early stage of the disease then shifts to Th-1 that proceed in the late stage, but both cytokines increased in CL patients in comparison to control group. The immune response of CL infection is possibly regulated by both Th-1 and Th-2. Multiple sores patients showed an increase of anti leishmanial IgE (0.120 ± 0.014), total IgE (120.7 ± 39.58 IU/ml), IFN-γ (87.4 ± 30.52 pg/ml) and IL-4 (63.70 ± 20.32 pg/ml) levels than single sore patients with mean value of 0.108 ± 0.14, 92.3 ± 35.23 IU/ml, 47.2 ± 27.80 pg/ml and 51.04 ± 15.0 pg/ml respectively. It can be presented also as ratio of INF-γ/IL-4 = 1.37 which is greater than those for single sore 0.9. These results indicated that the immune response of multiple sores patient's is higher than that with single sores.
Collapse
Affiliation(s)
- Ban Noori Al-Qadhi
- Biology Department, College of Science, University of Baghdad, Baghdad, Iraq
| | - Israa Salim Musa
- Biology Department, College of Science, University of Baghdad, Baghdad, Iraq
| | | |
Collapse
|
45
|
Gimblet C, Loesche MA, Carvalho L, Carvalho EM, Grice EA, Artis D, Scott P. IL-22 Protects against Tissue Damage during Cutaneous Leishmaniasis. PLoS One 2015; 10:e0134698. [PMID: 26285207 PMCID: PMC4540492 DOI: 10.1371/journal.pone.0134698] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/13/2015] [Indexed: 01/11/2023] Open
Abstract
Cutaneous leishmaniasis is a disease characterized by ulcerating skin lesions, the resolution of which requires an effective, but regulated, immune response that limits parasite growth without causing permanent tissue damage. While mechanisms that control the parasites have been well studied, the factors regulating immunopathologic responses are less well understood. IL-22, a member of the IL-10 family of cytokines, can contribute to wound healing, but in other instances promotes pathology. Here we investigated the role of IL-22 during leishmania infection, and found that IL-22 limits leishmania-induced pathology when a certain threshold of damage is induced by a high dose of parasites. Il22-/- mice developed more severe disease than wild-type mice, with significantly more pathology at the site of infection, and in some cases permanent loss of tissue. The increased inflammation was not due to an increased parasite burden, but rather was associated with the loss of a wound healing phenotype in keratinocytes. Taken together, these studies demonstrate that during cutaneous leishmaniasis, IL-22 can play a previously unappreciated role in controlling leishmania-induced immunopathology.
Collapse
Affiliation(s)
- Ciara Gimblet
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - Michael A. Loesche
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - Lucas Carvalho
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia—Doenças Tropicais, Salvador, Bahia, Brazil
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Edgar M. Carvalho
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia—Doenças Tropicais, Salvador, Bahia, Brazil
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Elizabeth A. Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - David Artis
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| |
Collapse
|
46
|
Rostamian M, Sohrabi S, Kavosifard H, Niknam HM. Lower levels of IgG1 in comparison with IgG2a are associated with protective immunity against Leishmania tropica infection in BALB/c mice. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2015; 50:160-166. [PMID: 26066544 DOI: 10.1016/j.jmii.2015.05.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 04/06/2015] [Accepted: 05/05/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND/PURPOSE Leishmania (L.) tropica is the causative agent of different forms of human leishmaniasis. There is little information about the role of Leishmania-specific antibodies in the immune response against L. tropica infection. The aim of this study is to evaluate the role of Leishmania-specific antibodies and their immunoglobulin G (IgG) isotypes in L. tropica infection. METHODS L. tropica at two different doses (high dose, 106 parasites/mouse and low dose, 103 parasites/mouse) were used for infection of BALB/c mice. BALB/c mice infected with Leishmania major were used for comparison. Anti-Leishmania antibodies of the IgG1 and IgG2a isotypes were assayed by enzyme-linked immunosorbent assay. RESULTS Our data showed that (1) a higher parasite dose results in higher levels of antibody. (2) L. tropica infection results in a lower IgG1 antibody response, compared with L. major infection. (3) The IgG2a/IgG1 antibody response in L. tropica infection is higher than that in L. major infection. CONCLUSION A higher IgG2a/IgG1 ratio is associated with protective immune response in L. tropica infection. These data can help to approach the complex profile of immunity against L. tropica infection.
Collapse
Affiliation(s)
| | - Saeed Sohrabi
- Department of Microbiology, Qom Branch of Islamic Azad University, Qom, Iran
| | - Hanie Kavosifard
- Department of Microbiology, Kurdistan Science and Research Branch, Islamic Azad University, Sanandaj, Iran
| | - Hamid M Niknam
- Immunology Department, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
47
|
Sun Y, Lu Y, Li T, Xie L, Deng Y, Li S, Qin X. Interferon Gamma +874T/A Polymorphism Increases the Risk of Hepatitis Virus-Related Diseases: Evidence from a Meta-Analysis. PLoS One 2015; 10:e0121168. [PMID: 25939029 PMCID: PMC4418602 DOI: 10.1371/journal.pone.0121168] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/28/2015] [Indexed: 02/06/2023] Open
Abstract
Background Interferon gamma (IFN-γ) is a key regulatory cytokine, which plays an important role in antiviral defense of an infected host. However, the association between the IFN-γ +874T/A gene polymorphism and hepatitis virus-related diseases is heterogeneous. Methods Based on the Preferred Reporting Items for Systematic Reviews and Meta-analyses statement, a comprehensive literature search of eligible studies in Embase, Pubmed, and the Cochrane Library was undertaken through November 2014. Odds ratios (ORs) and the corresponding 95% confidence intervals (CIs) were used to measure the strength of the models. Results Seventeen case-control articles, including 24 studies with 5503 individuals, met the inclusion criteria. The results indicated a statistically significant association between the IFN-γ +874T/A polymorphism and hepatitis virus—related diseases in a recessive gene model (AA vs. TT+TA: OR=1.350, 95% CI=1.101-1.657, P=0.004, I2%=54.3, and PQ=0.001 for heterogeneity), especially in Asians (OR=1.407, 95% CI=1.035-1.911, P=0.029, I2%=61.9, and PQ=0.005 for heterogeneity) and hepatitis B virus (HBV)–related disease (OR=1.486, 95% CI=1.195–1.849, P=0.000, I2%=40.4, and PQ=0.053 for heterogeneity). Conclusions The evidence suggests that the IFN-γ +874T/A polymorphism increases the risk of hepatitis virus—related diseases, especially in Asians and HBV—related diseases. Further studies on this topic in different ethnicities, especially genome-wide association studies, should be conducted to strengthen our results.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yu Lu
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Taijie Li
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Li Xie
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yan Deng
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Shan Li
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xue Qin
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- * E-mail:
| |
Collapse
|
48
|
Klose CSN, Diefenbach A. Transcription factors controlling innate lymphoid cell fate decisions. Curr Top Microbiol Immunol 2015; 381:215-55. [PMID: 25038936 DOI: 10.1007/82_2014_381] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mucosal epithelium is in direct contact with symbiotic and pathogenic microorganisms. Therefore, the mucosal surface is the principal portal of entry for invading pathogens and immune cells accumulated in the intestine to prevent infections. In addition to these conventional immune system functions, it has become clear that immune cells during steady-state continuously integrate microbial and nutrient-derived signals from the environment to support organ homeostasis. A major role in both processes is played by a recently discovered group of lymphocytes referred to as innate lymphoid cells (ILCs) Innate lymphoid cells (ILCs) that are specifically enriched at mucosal surfaces but are rather rare in secondary lymphoid organs. In analogy to the dichotomy between CD8 and CD4 T cells, we propose to classify ILCs into interleukin-7 receptor α-negative cytotoxic ILCs and IL-7Rα(+) helper-like ILCs. Dysregulated immune responses triggered by the various ILC subsets have been linked to inflammatory diseases such as inflammatory bowel disease, atopic dermatitis and airway hyperresponsiveness. Here, we will review recent progress in determining the transcriptional and developmental programs that control ILC fate decisions.
Collapse
Affiliation(s)
- Christoph S N Klose
- Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | | |
Collapse
|
49
|
Chowdhury BP, Bandyopadhyay S, Das S, Majumder S, Jha MK, Majumdar SB, Saha B, Majumdar S. The host-protective effect of arabinosylated lipoarabinomannan against Leishmania donovani infection is associated with restoration of IFN-γ responsiveness. PLoS One 2015; 10:e0117247. [PMID: 25658110 PMCID: PMC4319725 DOI: 10.1371/journal.pone.0117247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/22/2014] [Indexed: 12/20/2022] Open
Abstract
Visceral leishmaniasis (VL), which is endemic as a major infectious disease in the tropical and subtropical countries, is caused by a protozoan parasite Leishmania donovani. At present, restricted treatment options and lack of vaccines intensify the problem of controlling VL. Therefore, finding a novel immunoprophylactic or therapeutic principle is a pressing need. Here, we report that arabinosylated lipoarabinomannan (Ara-LAM), a TLR2-ligand isolated from Mycobacterium smegmatis, exhibits a strong immunomodulatory property that conferred protection against L. donovani infection. Although, Ara-LAM modulates TLR2 and MAPK signaling, it is not known whether Ara-LAM involves IFN-γ signaling for effective parasite clearance. Because, it is reported that IFN-γ signaling, a principle mediator of NO generation and macrophage and Tcell activation, is hampered during leishmanial pathogenesis. Ara-LAM increases IFN-γ receptor expression and potentiates IFN-γ receptor signaling through JAK-STAT pathway. Moreover, Ara-LAM reciprocally modulates IRF4 and IRF8 expression and reinstates anti-leishmanial Th1 response that eventuates in significantly reduced parasite load in spleen and liver of L. donovani-infected BALB/c mice. IFN-γRα silencing resulted in the suppression of these host-protective mechanisms affected by Ara-LAM. Thus, Ara-LAM-mediated restoration of IFN-γ responsiveness is a novel immuno-modulatory principle for protection against L. donovani susceptible host.
Collapse
Affiliation(s)
| | | | - Shibali Das
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Saikat Majumder
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Mukesh Kumar Jha
- Laboratory-V, National Centre for Cell Science (NCCS), Pune, Maharashtra, India
| | | | - Bhaskar Saha
- Laboratory-V, National Centre for Cell Science (NCCS), Pune, Maharashtra, India
| | - Subrata Majumdar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
- * E-mail:
| |
Collapse
|
50
|
Sun Y, Lu Y, Pen Q, Li T, Xie L, Deng Y, Qin A. Interferon gamma +874 T/A polymorphism increases the risk of cervical cancer: evidence from a meta-analysis. Tumour Biol 2015; 36:4555-64. [PMID: 25649976 DOI: 10.1007/s13277-015-3100-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/01/2014] [Indexed: 02/02/2023] Open
Abstract
Data from previous studies about the association between interferon gamma (IFN-γ) +874 T/A (rs2430561) polymorphism and cervical cancer risk offer controversial results. To obtain a more dependable conclusion, this meta-analysis was performed. We selected eight articles including nine case-control studies with 1,116 cases and 1,290 controls, odds ratios (OR) with 95 % confidence intervals (CI) were used to assess the strength of the association. Subgroup analysis was carried out by ethnicity, source of controls, genotyping methods, and score of quality assessment. Our meta-analysis indicated that the IFN-γ (+874 T/A) polymorphism significantly increased the risk of cervical cancer in the codominant model (TA vs. TT: OR = 1.471, 95 % CI = 1.137-1.903, P = 0.003, I (2) % = 0.0, P Q = 0.785) and the dominant model (TA + AA vs. TT: OR = 1.399, 95 % CI = 1.097-1.784, P = 0.007, I (2) % = 0.0, P Q = 0.486) in the overall population. Stratified analysis by ethnicity indicated a significantly increased risk of cervical cancer in Asians in the codominant model (TA vs. TT: OR = 1.494, 95 % CI = 1.069-2.087, P = 0.019, I (2) % = 0.0, P Q = 0.440) and the dominant model (OR = 1.455, 95 % CI = 1.062-1.993, P = 0.019, I (2) % = 42.9, P Q = 0.154). Thus, the IFN-γ (+874 T/A) polymorphism is likely to increase the risk of cervical cancer. Because of the limited studies and sample sizes included in our meta-analysis, further well-designed and large-scale studies are demanded to confirm our results.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|