1
|
Yadav N, Parthiban C, Billman ZP, Stone BC, Watson FN, Zhou K, Olsen TM, Cruz Talavera I, Seilie AM, Kalata AC, Matsubara J, Shears MJ, Reynolds RA, Murphy SC. More time to kill: A longer liver stage increases T cell-mediated protection against pre-erythrocytic malaria. iScience 2023; 26:108489. [PMID: 38162031 PMCID: PMC10755051 DOI: 10.1016/j.isci.2023.108489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 10/16/2023] [Accepted: 11/14/2023] [Indexed: 01/03/2024] Open
Abstract
Liver stage (LS) Plasmodia mature in 2-2.5 days in rodents compared to 5-6 days in humans. Plasmodium-specific CD8+ T cell expansion differs across these varied timespans. To mimic the kinetics of CD8+ T cells of human Plasmodium infection, a two-dose challenge mouse model that achieved 4-5 days of LS antigen exposure was developed. In this model, mice were inoculated with a non-protective, low dose of late-arresting, genetically attenuated sporozoites to initiate T cell activation and then re-inoculated 2-3 days later with wild-type sporozoites. Vaccines that partially protected against traditional challenge completely protected against two-dose challenge. During the challenge period, CD8+ T cell frequencies increased in the livers of two-dose challenged mice but not in traditionally challenged mice, further suggesting that this model better recapitulates kinetics of CD8+ T cell expansion in humans during the P. falciparum LS. Vaccine development and antigen discovery efforts may be aided by using the two-dose challenge strategy.
Collapse
Affiliation(s)
- Naveen Yadav
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Chaitra Parthiban
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Zachary P. Billman
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Brad C. Stone
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Felicia N. Watson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Kevin Zhou
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Tayla M. Olsen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Irene Cruz Talavera
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Annette Mariko Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Anya C. Kalata
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Jokichi Matsubara
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Melanie J. Shears
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Rebekah A. Reynolds
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Joyce S, Okoye GD, Driver JP. Die Kämpfe únd schláchten-the struggles and battles of innate-like effector T lymphocytes with microbes. Front Immunol 2023; 14:1117825. [PMID: 37168859 PMCID: PMC10165076 DOI: 10.3389/fimmu.2023.1117825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/22/2023] [Indexed: 05/13/2023] Open
Abstract
The large majority of lymphocytes belong to the adaptive immune system, which are made up of B2 B cells and the αβ T cells; these are the effectors in an adaptive immune response. A multitudinous group of lymphoid lineage cells does not fit the conventional lymphocyte paradigm; it is the unconventional lymphocytes. Unconventional lymphocytes-here called innate/innate-like lymphocytes, include those that express rearranged antigen receptor genes and those that do not. Even though the innate/innate-like lymphocytes express rearranged, adaptive antigen-specific receptors, they behave like innate immune cells, which allows them to integrate sensory signals from the innate immune system and relay that umwelt to downstream innate and adaptive effector responses. Here, we review natural killer T cells and mucosal-associated invariant T cells-two prototypic innate-like T lymphocytes, which sense their local environment and relay that umwelt to downstream innate and adaptive effector cells to actuate an appropriate host response that confers immunity to infectious agents.
Collapse
Affiliation(s)
- Sebastian Joyce
- Department of Veterans Affairs, Tennessee Valley Healthcare Service, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, The Vanderbilt Institute for Infection, Immunology and Inflammation and Vanderbilt Center for Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Gosife Donald Okoye
- Department of Pathology, Microbiology and Immunology, The Vanderbilt Institute for Infection, Immunology and Inflammation and Vanderbilt Center for Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - John P. Driver
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
3
|
Deng S, Graham ML, Chen XM. The Complexity of Interferon Signaling in Host Defense against Protozoan Parasite Infection. Pathogens 2023; 12:319. [PMID: 36839591 PMCID: PMC9962834 DOI: 10.3390/pathogens12020319] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Protozoan parasites, such as Plasmodium, Leishmania, Toxoplasma, Cryptosporidium, and Trypanosoma, are causative agents of health-threatening diseases in both humans and animals, leading to significant health risks and socioeconomic losses globally. The development of effective therapeutic and prevention strategies for protozoan-caused diseases requires a full understanding of the pathogenesis and protective events occurring in infected hosts. Interferons (IFNs) are a family of cytokines with diverse biological effects in host antimicrobial defense and disease pathogenesis, including protozoan parasite infection. Type II IFN (IFN-γ) has been widely recognized as the essential defense cytokine in intracellular protozoan parasite infection, whereas recent studies also revealed the production and distinct function of type I and III IFNs in host defense against these parasites. Decoding the complex network of the IFN family in host-parasite interaction is critical for exploring potential new therapeutic strategies against intracellular protozoan parasite infection. Here, we review the complex effects of IFNs on the host defense against intracellular protozoan parasites and the crosstalk between distinct types of IFN signaling during infections.
Collapse
Affiliation(s)
- Silu Deng
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Marion L. Graham
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Xian-Ming Chen
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
4
|
Alizadeh Z, Omidnia P, Altalbawy FMA, Gabr GA, Obaid RF, Rostami N, Aslani S, Heidari A, Mohammadi H. Unraveling the role of natural killer cells in leishmaniasis. Int Immunopharmacol 2023; 114:109596. [PMID: 36700775 DOI: 10.1016/j.intimp.2022.109596] [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: 10/06/2022] [Revised: 12/07/2022] [Accepted: 12/11/2022] [Indexed: 12/24/2022]
Abstract
NK cells are known as frontline responders that are efficient in combating several maladies as well as leishmaniasis caused by Leishmania spp. As such they are being investigated to be used for adoptive transfer therapy and vaccine. In spite of the lack of antigen-specific receptors at their surface, NK cells can selectively recognize pathogens, accomplished by the activation of the receptors on the NK cell surface and also as the result of their effector functions. Activation of NK cells can occur through interaction between TLR-2 expressed on NK cells and. LPG of Leishmania parasites. In addition, NK cell activation can occur by cytokines (e.g., IFN-γ and IL-12) that also lead to producing cytokines and chemokines and lysis of target cells. This review summarizes several evidences that support NK cells activation for controlling leishmaniasis and the potentially lucrative roles of NK cells during leishmaniasis. Furthermore, we discuss strategies of Leishmania parasites in inhibiting NK cell functions. Leishmania LPG can utilizes TLR2 to evade host-immune responses. Also, Leishmania GP63 can directly binds to NK cells and modulates NK cell phenotype. Finally, this review analyzes the potentialities to harness NK cells effectiveness in therapy regimens and vaccinations.
Collapse
Affiliation(s)
- Zahra Alizadeh
- Department of Parasitology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Farag M A Altalbawy
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza 12613, Egypt; Department of Chemistry, University College of Duba, University of Tabuk, Duba 71911, Saudi Arabia
| | - Gamal A Gabr
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center, Giza, Egypt
| | - Rasha Fadhel Obaid
- Department of Biomedical Engineering, Al-Mustaqbal University College, Babylon, Iraq
| | - Narges Rostami
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliehsan Heidari
- Department of Parasitology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
5
|
Gálvez NMS, Bohmwald K, Pacheco GA, Andrade CA, Carreño LJ, Kalergis AM. Type I Natural Killer T Cells as Key Regulators of the Immune Response to Infectious Diseases. Clin Microbiol Rev 2021; 34:e00232-20. [PMID: 33361143 PMCID: PMC7950362 DOI: 10.1128/cmr.00232-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The immune system must work in an orchestrated way to achieve an optimal response upon detection of antigens. The cells comprising the immune response are traditionally divided into two major subsets, innate and adaptive, with particular characteristics for each type. Type I natural killer T (iNKT) cells are defined as innate-like T cells sharing features with both traditional adaptive and innate cells, such as the expression of an invariant T cell receptor (TCR) and several NK receptors. The invariant TCR in iNKT cells interacts with CD1d, a major histocompatibility complex class I (MHC-I)-like molecule. CD1d can bind and present antigens of lipid nature and induce the activation of iNKT cells, leading to the secretion of various cytokines, such as gamma interferon (IFN-γ) and interleukin 4 (IL-4). These cytokines will aid in the activation of other immune cells following stimulation of iNKT cells. Several molecules with the capacity to bind to CD1d have been discovered, including α-galactosylceramide. Likewise, several molecules have been synthesized that are capable of polarizing iNKT cells into different profiles, either pro- or anti-inflammatory. This versatility allows NKT cells to either aid or impair the clearance of pathogens or to even control or increase the symptoms associated with pathogenic infections. Such diverse contributions of NKT cells to infectious diseases are supported by several publications showing either a beneficial or detrimental role of these cells during diseases. In this article, we discuss current data relative to iNKT cells and their features, with an emphasis on their driving role in diseases produced by pathogenic agents in an organ-oriented fashion.
Collapse
Affiliation(s)
- Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaspar A Pacheco
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leandro J Carreño
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
6
|
Keswani T, Delcroix-Genete D, Herbert F, Leleu I, Lambert C, Draheim M, Salome-Desnoulez S, Saliou JM, Cazenave PA, Silvie O, Roland J, Pied S. Plasmodium yoelii Uses a TLR3-Dependent Pathway to Achieve Mammalian Host Parasitism. THE JOURNAL OF IMMUNOLOGY 2020; 205:3071-3082. [PMID: 33148715 DOI: 10.4049/jimmunol.1901317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Malaria is associated with complicated immunopathogenesis. In this study, we provide evidence for an unexpected role of TLR3 in promoting the establishment of Plasmodium yoelii infection through delayed clearance of parasitemia in wild type C57BL/6jRj (B6) compared with TLR3 knockout mice. In this study, we confirmed an increased expression of Tlr3, Trif, Tbk1, and Irf7/Irf3 in the liver 42 h postinfection and the initiation of an early burst of proinflammatory response such as Ifng, NF-kB, and Tnfa in B6 mice that may promote parasite fitness. Interestingly, in the absence of TLR3, we showed the involvement of high IFN-γ and lower type I IFN response in the early clearance of parasitemia. In parallel, we observed an increase in splenic NK and NKT cells expressing TLR3 in infected B6 mice, suggesting a role for TLR sensing in the innate immune response. Finally, we find evidence that the increase in the frequency of CD19+TLR3+ B cells along with reduced levels of total IgG in B6 mice possibly suggests the initiation of TLR3-dependent pathway early during P. yoelii infection. Our results thus reveal a new mechanism in which a parasite-activated TLR3 pathway promotes blood stage infection along with quantitative and qualitative differences in Ab responses.
Collapse
Affiliation(s)
- Tarun Keswani
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Delphine Delcroix-Genete
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Fabien Herbert
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Ines Leleu
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Claire Lambert
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Marion Draheim
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | | | - Jean Michel Saliou
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Pierre-André Cazenave
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Olivier Silvie
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, 75013 Paris, France
| | - Jacques Roland
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Sylviane Pied
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France;
| |
Collapse
|
7
|
Ng SS, Engwerda CR. Innate Lymphocytes and Malaria - Players or Spectators? Trends Parasitol 2018; 35:154-162. [PMID: 30579700 DOI: 10.1016/j.pt.2018.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/29/2018] [Accepted: 11/29/2018] [Indexed: 12/19/2022]
Abstract
Malaria remains an important global disease. Despite significant advances over the past decade in reducing disease morbidity and mortality, new measures are needed if malaria is to be eliminated. Significant advances in our understanding about host immune responses during malaria have been made, opening up opportunities to generate long-lasting antiparasitic immunity through vaccination or immune therapy. However, there is still much debate over which immune cell populations contribute to immunity to malaria, including innate lymphocytes that comprise recently identified innate lymphoid cells (ILCs) and better known innate-like T cell subsets. Here, we review research on these immune cell subsets and discuss whether they have any important roles in immunity to malaria or if they are redundant.
Collapse
Affiliation(s)
- Susanna S Ng
- Immunology and Infection Laboratory, QIMR Berghofer Medical Research Institute, QLD, Australia; School of Environment and Science, Griffith University, QLD, Australia
| | - Christian R Engwerda
- Immunology and Infection Laboratory, QIMR Berghofer Medical Research Institute, QLD, Australia.
| |
Collapse
|
8
|
Wikenheiser DJ, Brown SL, Lee J, Stumhofer JS. NK1.1 Expression Defines a Population of CD4 + Effector T Cells Displaying Th1 and Tfh Cell Properties That Support Early Antibody Production During Plasmodium yoelii Infection. Front Immunol 2018; 9:2277. [PMID: 30374346 PMCID: PMC6196288 DOI: 10.3389/fimmu.2018.02277] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/13/2018] [Indexed: 11/23/2022] Open
Abstract
Early plasmablast induction is a hallmark of Plasmodium infection and is thought to contribute to the control of acute parasite burden. Although long understood to be a T-cell dependent phenomenon, regulation of early plasmablast differentiation, however, is poorly understood. Here, we identify a population of CD4+ T cells that express the innate NK cell marker NK1.1 as an important source of T cell help for early plasmablast and parasite-specific Ab production. Interestingly, NK1.1+ CD4+ T cells arise from conventional, naive NK1.1− CD4+ T cells, and their generation is independent of CD1d but critically reliant on MHC-II. CD4+ T cells that express NK1.1 early after activation produce IFN-γ and IL-21, and express the follicular helper T (Tfh) cell markers ICOS, PD-1 and CXCR5 more frequently than NK1.1− CD4+ T cells. Further analysis of this population revealed that NK1.1+ Tfh-like cells were more regularly complexed with plasmablasts than NK1.1− Tfh-like cells. Ultimately, depletion of NK1.1+ cells impaired class-switched parasite-specific antibody production during early Plasmodium yoelii infection. Together, these data suggest that expression of NK1.1 defines a population of rapidly expanding effector CD4+ T cells that specifically promote plasmablast induction during Plasmodium infection and represent a subset of T cells whose modulation could promote effective vaccine design.
Collapse
Affiliation(s)
- Daniel J Wikenheiser
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Susie L Brown
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Juhyung Lee
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
9
|
Silvie O, Amino R, Hafalla JC. Tissue-specific cellular immune responses to malaria pre-erythrocytic stages. Curr Opin Microbiol 2017; 40:160-167. [PMID: 29217460 DOI: 10.1016/j.mib.2017.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 11/30/2022]
Abstract
Complete and long-lasting protective immunity against malaria can be achieved through vaccination with invasive live attenuated Plasmodium sporozoites, the motile stage inoculated in the host skin during a mosquito bite. Protective immunity relies primarily on effector CD8+ T cells targeting the parasite in the liver. Understanding the tissue-specific features of the immune response is emerging as a vital requirement for understanding protective immunity. The small parasite inoculum, the scarcity of infected cells and the tolerogenic properties of the liver represent hurdles for the establishment of protective immunity in endemic areas. In this review, we discuss recent advances on liver-specific features of immunity including innate recognition of malaria pre-erythrocytic stages, CD8+ T cell interactions with infected hepatocytes, antigen presentation for effective CD8+ T cell responses and generation of liver-resident memory CD8+ T cells. A better understanding of the factors involved in the induction and maintenance of effector CD8+ T cell immunity against malaria pre-erythrocytic stages is crucial for the development of an effective vaccine targeting the initial phase of malaria infection.
Collapse
Affiliation(s)
- Olivier Silvie
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, Paris, France.
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.
| | - Julius Clemence Hafalla
- Immunology and Infection Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| |
Collapse
|
10
|
Pinna RA, Silva-Dos-Santos D, Perce-da-Silva DS, Oliveira-Ferreira J, Villa-Verde DMS, De Luca PM, Banic DM. Malaria-Cutaneous Leishmaniasis Co-infection: Influence on Disease Outcomes and Immune Response. Front Microbiol 2016; 7:982. [PMID: 27446022 PMCID: PMC4921482 DOI: 10.3389/fmicb.2016.00982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/07/2016] [Indexed: 12/16/2022] Open
Abstract
Malaria and Cutaneous Leishmaniasis (CL) are co-endemic throughout large regions in tropical countries and co-infection may impact the evolution of host-parasite interactions. In the present study, we evaluate Malaria/Leishmaniasis disease outcome, Th1/Th2 cytokine levels and the CD4 and CD8 T-cell profiles in a co-infection murine model (BALB/c) of Plasmodium yoelii 17XNL (Py) and Leishmania amazonensis (La) or L. braziliensis (Lb). Malaria parasitaemia was assessed through blood strains stained with Giemsa. Leishmania lesions were monitored with a digital caliper and parasite loads determined by limiting-dilution assay. Serum levels of IFN-γ, TNF, IL-2, IL-4, IL-6, IL-10, and IL-17 were determined using multiplexed bead assay and expression of CD3, CD4, and CD8 T-cells markers were determined by Flow Cytometry in the thymus, spleens and lymph nodes. Parasitaemia in Lb+Py co-infected group was lower than in Py single-infected group, suggesting a protective effect of Lb co-infection in Malaria progression. In contrast, La+Py co-infection increased parasitaemia, patent infection and induced mortality in non-lethal Malaria infection. Regarding Leishmaniasis, Lb+Py co-infected group presented smaller lesions and less ulceration than Lb single-infected animals. In contrast, La+Py co-infected group presented only a transitory delay on the development of lesions when compared to La single-infected mice. Decreased levels of IFN-γ, TNF, IL-6, and IL-10 were observed in the serum of co-infected groups, demonstrating a modulation of Malaria immune response by Leishmania co-infections. We observed an intense thymic atrophy in Py single-infected and co-infected groups, which recovered earlier in co-infected animals. The CD4 and CD8 T cell profiles in thymus, spleens and lymph nodes did not differ between Py single and co-infected groups, except for a decrease in CD4+CD8+ T cells which also increased faster in co-infected mice. Our results suggest that Py and Leishmania co-infection may change disease outcome. Interestingly Malaria outcome can be altered according to the Leishmania specie involved. Alternatively Malaria infection reduced the severity or delayed the onset of leishmanial lesions. These alterations in Malaria and CL development seem to be closely related with changes in the immune response as demonstrated by alteration in serum cytokine levels and thymus/spleens T cell phenotypes dynamics during infection.
Collapse
Affiliation(s)
- Raquel A Pinna
- Laboratory of Simulids, Onchocerciasis and Sympatric Diseases: Mansonelliasis and Malaria, Oswaldo Cruz Institute, Oswaldo Cruz Foundation Rio de Janeiro, Brazil
| | - Danielle Silva-Dos-Santos
- Laboratory of Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation Rio de Janeiro, Brazil
| | - Daiana S Perce-da-Silva
- Laboratory of Simulids, Onchocerciasis and Sympatric Diseases: Mansonelliasis and Malaria, Oswaldo Cruz Institute, Oswaldo Cruz Foundation Rio de Janeiro, Brazil
| | - Joseli Oliveira-Ferreira
- Laboratory of Immunoparasitology Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation Rio de Janeiro, Brazil
| | - Dea M S Villa-Verde
- Laboratory of Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation Rio de Janeiro, Brazil
| | - Paula M De Luca
- Laboratory of Immunoparasitology Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation Rio de Janeiro, Brazil
| | - Dalma M Banic
- Laboratory of Simulids, Onchocerciasis and Sympatric Diseases: Mansonelliasis and Malaria, Oswaldo Cruz Institute, Oswaldo Cruz Foundation Rio de Janeiro, Brazil
| |
Collapse
|
11
|
The TLR2 is activated by sporozoites and suppresses intrahepatic rodent malaria parasite development. Sci Rep 2015; 5:18239. [PMID: 26667391 PMCID: PMC4678895 DOI: 10.1038/srep18239] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/24/2015] [Indexed: 01/08/2023] Open
Abstract
TLRs (Toll-like receptors) play an important role in the initiation of innate immune responses against invading microorganisms. Although several TLRs have been reported to be involved in the innate immune response against the blood-stage of malaria parasites, the role of TLRs in the development of the pre-erythrocytic stage is still largely unknown. Here, we found that sporozoite and its lysate could significantly activate the TLR2, and induce macrophages to release proinflammatory cytokines, including IL-6, MCP-1 and TNF-α, in a TLR2-dependent manner. Further studies showed that sporozoite and its lysate could be recognized by either TLR2 homodimers or TLR2/1 and TLR2/6 heterodimers, implicating the complexity of TLR2 agonist in sporozoite. Interestingly, the TLR2 signaling can significantly suppress the development of the pre-erythrocytic stage of Plasmodium yoelii, as both liver parasite load and subsequent parasitemia were significantly elevated in both TLR2- and MyD88-deficient mice. Additionally, the observed higher level of parasite burden in TLR2−/− mice was found to be closely associated with a reduction in proinflammatory cytokines in the liver. Therefore, we provide the first evidence that sporozoites can activate the TLR2 signaling, which in turn significantly inhibits the intrahepatic parasites. This may provide us with novel clues to design preventive anti-malaria therapies.
Collapse
|
12
|
Van Kaer L, Parekh VV, Wu L. The Response of CD1d-Restricted Invariant NKT Cells to Microbial Pathogens and Their Products. Front Immunol 2015; 6:226. [PMID: 26029211 PMCID: PMC4429631 DOI: 10.3389/fimmu.2015.00226] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/27/2015] [Indexed: 12/18/2022] Open
Abstract
Invariant natural killer T (iNKT) cells become activated during a wide variety of infections. This includes organisms lacking cognate CD1d-binding glycolipid antigens recognized by the semi-invariant T cell receptor of iNKT cells. Additional studies have shown that iNKT cells also become activated in vivo in response to microbial products such as bacterial lipopolysaccharide, a potent inducer of cytokine production in antigen-presenting cells (APCs). Other studies have shown that iNKT cells are highly responsive to stimulation by cytokines such as interleukin-12. These findings have led to the concept that microbial pathogens can activate iNKT cells either directly via glycolipids or indirectly by inducing cytokine production in APCs. iNKT cells activated in this manner produce multiple cytokines that can influence the outcome of infection, usually in favor of the host, although potent iNKT cell activation may contribute to an uncontrolled cytokine storm and sepsis. One aspect of the response of iNKT cells to microbial pathogens is that it is short-lived and followed by an extended time period of unresponsiveness to reactivation. This refractory period may represent a means to avoid chronic activation and cytokine production by iNKT cells, thus protecting the host against some of the negative effects of iNKT cell activation, but potentially putting the host at risk for secondary infections. These effects of microbial pathogens and their products on iNKT cells are not only important for understanding the role of these cells in immune responses against infections but also for the development of iNKT cell-based therapies.
Collapse
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine , Nashville, TN , USA
| | - Vrajesh V Parekh
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine , Nashville, TN , USA
| | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine , Nashville, TN , USA
| |
Collapse
|
13
|
Lactobacillus casei CRL 431 administration decreases inflammatory cytokines in a diet-induced obese mouse model. Nutrition 2015; 31:1000-7. [PMID: 26059375 DOI: 10.1016/j.nut.2015.02.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 02/04/2015] [Accepted: 02/17/2015] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Obesity is a chronic disease associated with an inflammatory process in which cytokines play an important role. Probiotic microorganisms have been associated with modulation of the host immune system. The aim of this study was to evaluate the influence of the probiotic bacterium Lactobacillus casei CRL 431 on the cytokine response in a model of mice under high-fat diet (HFD) conditions. METHODS BALB/c mice received a conventional balanced diet or an HFD. The test groups received milk, milk fermented by L. casei (FM), or L. casei as suspension in the drinking water. Proinflammatory and regulatory cytokine producer cells were evaluated in the small intestine and liver; the cytokine levels in the intestinal fluids were also evaluated. The percentages of immune cells as macrophages (F4/80), NKT, CD4+, CD8+ populations were determined in the liver. Adipocytes were also isolated and cultured to evaluate cytokines and the chemokine monocyte chemoattractant protein (MCP)-1 produced by them. RESULTS The administration of probiotic L. casei CRL 431 exerted an anti-inflammatory response in mice fed an HFD, evidenced mainly by decreasing proinflammatory cytokines, such as interleukin (IL)-6, IL-17, and tumor necrosis factor-α. Probiotic administration also was associated with fewer immune-infiltrating cells in the liver of mice that received the HFD and decreased secretion of MCP-1 by the adipocytes. This last observation could be associated with less macrophage accumulation in the adipose tissues, which is characteristic in the obese host and contributes to maintaining the inflammatory response in this organ. The results obtained show an anti-inflammatory effect of L. casei CRL 431 when it is administered as a supplement of the HFD in a mouse model.
Collapse
|
14
|
Miller J, Sack B, Baldwin M, Vaughan A, Kappe S. Interferon-Mediated Innate Immune Responses against Malaria Parasite Liver Stages. Cell Rep 2014; 7:436-447. [DOI: 10.1016/j.celrep.2014.03.018] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 02/07/2014] [Accepted: 03/11/2014] [Indexed: 01/25/2023] Open
|
15
|
Cheng Q, Zhang Q, Xu X, Yin L, Sun L, Lin X, Dong C, Pan W. MAPK phosphotase 5 deficiency contributes to protection against blood-stage Plasmodium yoelii 17XL infection in mice. THE JOURNAL OF IMMUNOLOGY 2014; 192:3686-96. [PMID: 24634491 DOI: 10.4049/jimmunol.1301863] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell-mediated immunity plays a crucial role in the development of host resistance to asexual blood-stage malaria infection. However, little is known of the regulatory factors involved in this process. In this study, we investigated the impact of MAPK phosphotase 5 (MKP5) on protective immunity against a lethal Plasmodium yoelii 17XL blood-stage infection using MKP5 knockout C57BL/6 mice. Compared with wild-type control mice, MKP5 knockout mice developed significantly lower parasite burdens with prolonged survival times. We found that this phenomenon correlated with a rapid and strong IFN-γ-dependent cellular immune response during the acute phase of infection. Inactivation of IFN-γ by the administration of a neutralizing Ab significantly reduced the protective effects in MKP5 knockout mice. By analyzing IFN-γ production in innate and adaptive lymphocyte subsets, we observed that MKP5 deficiency specifically enhanced the IFN-γ response mediated by CD4+ T cells, which was attributable to the increased stimulatory capacity of splenic CD11c+ dendritic cells. Furthermore, following vaccination with whole blood-stage soluble plasmodial Ag, MKP5 knockout mice acquired strongly enhanced Ag-specific immune responses and a higher level of protection against subsequent P. yoelii 17XL challenge. Finally, we found the enhanced response mediated by MKP5 deficiency resulted in a lethal consequence in mice when infected with nonlethal P. yoelii 17XNL. Thus, our data indicate that MKP5 is a potential regulator of immune resistance against Plasmodium infection in mice, and that an understanding of the role of MKP5 in manipulating anti-malaria immunity may provide valuable information on the development of better control strategies for human malaria.
Collapse
Affiliation(s)
- Qianqian Cheng
- Institute of Infectious Disease and Vaccine Development, Tongji University School of Medicine, Shanghai 200092, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Adachi K, Osada Y, Nakamura R, Tamada K, Hamano S. Unique T cells with unconventional cytokine profiles induced in the livers of mice during Schistosoma mansoni infection. PLoS One 2013; 8:e82698. [PMID: 24358216 PMCID: PMC3865148 DOI: 10.1371/journal.pone.0082698] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/26/2013] [Indexed: 12/31/2022] Open
Abstract
During infection with Schistosoma, serious hepatic disorders are induced in the host. The liver possesses unique immune systems composed of specialized cells that differ from those of other immune competent organs or tissues. Host immune responses change dramatically during Schistosoma mansoni infection; in the early phase, Th1-related responses are induced, whereas during the late phase Th2 reactions dominate. Here, we describe unique T cell populations induced in the liver of mice during the period between Th1- and Th2-phases, which we term the transition phase. During this phase, varieties of immune cells including T lymphocytes increase in the liver. Subsets of CD4+ T cells exhibit unique cytokine production profiles, simultaneously producing both IFN-γ and IL-13 or both IFN-γ and IL-4. Furthermore, cells triply positive for IFN-γ, IL-13 and IL-4 also expand in the S. mansoni-infected liver. The induction of these unique cell populations does not occur in the spleen, indicating it is a phenomenon specific to the liver. In single hepatic CD4+ T cells showing the unique cytokine profiles, both T-bet and GATA-3 are expressed. Thus, our studies show that S. mansoni infection triggers the induction of hepatic T cell subsets with unique cytokine profiles.
Collapse
Affiliation(s)
- Keishi Adachi
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- Global Center of Excellence Program, Nagasaki University, Nagasaki, Japan
| | - Yoshio Osada
- Department of Immunology and Parasitology, The University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Japan
| | - Risa Nakamura
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Koji Tamada
- Department of Immunology and Cell Signaling Analysis, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- Global Center of Excellence Program, Nagasaki University, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
17
|
HLA class II (DR0401) molecules induce Foxp3+ regulatory T cell suppression of B cells in Plasmodium yoelii strain 17XNL malaria. Infect Immun 2013; 82:286-97. [PMID: 24166949 DOI: 10.1128/iai.00272-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Unlike human malaria parasites that induce persistent infection, some rodent malaria parasites, like Plasmodium yoelii strain 17XNL (Py17XNL), induce a transient (self-curing) malaria infection. Cooperation between CD4 T cells and B cells to produce antibodies is thought to be critical for clearance of Py17XNL parasites from the blood, with major histocompatibility complex (MHC) class II molecules being required for activation of CD4 T cells. In order to better understand the correspondence between murine malaria models and human malaria, and in particular the role of MHC (HLA) class II molecules, we studied the ability of humanized mice expressing human HLA class II molecules to clear Py17XNL infection. We showed that humanized mice expressing HLA-DR4 (DR0401) molecules and lacking mouse MHC class II molecules (EA(0)) have impaired production of specific antibodies to Py17XNL and cannot cure the infection. In contrast, mice expressing HLA-DR4 (DR0402), HLA-DQ6 (DQ0601), HLA-DQ8 (DQ0302), or HLA-DR3 (DR0301) molecules in an EA(0) background were able to elicit specific antibodies and self-cure the infection. In a series of experiments, we determined that the inability of humanized DR0401.EA(0) mice to elicit specific antibodies was due to expansion and activation of regulatory CD4(+) Foxp3(+) T cells (Tregs) that suppressed B cells to secrete antibodies through cell-cell interactions. Treg depletion allowed the DR0401.EA(0) mice to elicit specific antibodies and self-cure the infection. Our results demonstrated a differential role of MHC (HLA) class II molecules in supporting antibody responses to Py17XNL malaria and revealed a new mechanism by which malaria parasites stimulate B cell-suppressogenic Tregs that prevent clearance of infection.
Collapse
|
18
|
Razakandrainibe R, Pelleau S, Grau GE, Jambou R. Antigen presentation by endothelial cells: what role in the pathophysiology of malaria? Trends Parasitol 2012; 28:151-60. [PMID: 22365903 DOI: 10.1016/j.pt.2012.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 01/20/2012] [Accepted: 01/20/2012] [Indexed: 11/19/2022]
Abstract
Disruption of the endothelial cell (EC) barrier leads to pathology via edema and inflammation. During infections, pathogens are known to invade the EC barrier and modulate vascular permeability. However, ECs are semi-professional antigen-presenting cells, triggering T-cell costimulation and specific immune-cell activation. This in turn leads to the release of inflammatory mediators and the destruction of infected cells by effectors such as CD8(+) T-cells. During malaria, transfer of parasite antigens to the EC surface is now established. At the same time, CD8 activation seems to play a major role in cerebral malaria. We summarize here some of the pathways leading to antigen presentation by ECs and address the involvement of these mechanisms in the pathophysiology of cerebral malaria.
Collapse
|
19
|
Renna MS, Figueredo CM, Rodríguez-Galán MC, Icely PA, Peralta Ramos JM, Correa SG, Sotomayor CE. Abrogation of spontaneous liver tolerance during immune response to Candida albicans: contribution of NKT and hepatic mononuclear cells. Int Immunol 2012; 24:315-25. [DOI: 10.1093/intimm/dxs001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
20
|
Duwaerts CC, Gregory SH. Targeting the diverse immunological functions expressed by hepatic NKT cells. Expert Opin Ther Targets 2011; 15:973-88. [PMID: 21564001 DOI: 10.1517/14728222.2011.584874] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION NKT cells comprise approximately 30% of the hepatic lymphoid population in mice (∼ 50% in humans). Most mouse hepatic NKT cells [invariant (i)NKT cells] express T cell receptors, composed of invariant Vα14Jα18 chains. Unlike conventional T cells, iNKT cells recognize glycolipids presented in association with MHC class Ib (CD1d) molecules. Purportedly, iNKT cells serve key functions in several immunological events; the nature of these is often unclear. The consequences of hepatic iNKT cell activation can be beneficial or detrimental. α-Galactosylceramide stimulates the production of IFN-γ and IL-4. The reciprocal suppression exhibited by these cytokines limits the potential therapeutic value of α-galactosylceramide. Efforts are ongoing to develop α-galactosylceramide analogs that modulate iNKT cell activity and selectively promote IFN-γ or IL-4. AREAS COVERED An overview of hepatic iNKT cells and their purported role in liver disease. Efforts to develop therapeutic agents that promote their beneficial contributions. EXPERT OPINION While a growing body of literature documents the differential effects of α-GalCer analogs on IFN-γ and IL-4 production, the effects of these analogs on other iNKT cell activities remain to be determined. An exhaustive examination of the effects of these analogs on inflammation and liver injury in animal models remains prior to considering their utility in clinical trials.
Collapse
Affiliation(s)
- Caroline C Duwaerts
- Rhode Island Hospital and The Warren Alpert Medical School at Brown University, Department of Medicine, Providence, RI 02903, USA
| | | |
Collapse
|
21
|
Muxel SM, Freitas do Rosário AP, Sardinha LR, Castillo-Méndez SI, Zago CA, Rodriguez-Málaga SM, Alvarez Mosig JM, D'Império Lima MR. Comparative analysis of activation phenotype, proliferation, and IFN-gamma production by spleen NK1.1(+) and NK1.1(-) T cells during Plasmodium chabaudi AS malaria. J Interferon Cytokine Res 2010; 30:417-26. [PMID: 20187775 DOI: 10.1089/jir.2009.0095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The NK1.1 molecule participates in NK, NKT, and T-cell activation, contributing to IFN-gamma production and cytotoxicity. To characterize the early immune response to Plasmodium chabaudi AS, spleen NK1.1(+) and NK1.1(-) T cells were compared in acutely infected C57BL/6 mice. The first parasitemia peak in C57BL/6 mice correlated with increase in CD4(+)NK1.1(+)TCR-alphabeta(+), CD8(+)NK1.1(+)TCR-alphabeta(+), and CD4(+)NK1.1(-)TCR-alphabeta(+) cell numbers per spleen, where a higher increment was observed for NK1.1(+) T cells compared to NK1.1(-) T cells. According to the ability to recognize the CD1d-alpha-GalCer tetramer, CD4(+)NK1.1(+) cells in 7-day infected mice were not predominantly invariant NKT cells. At that time, nearly all NK1.1(+) T cells and around 30% of NK1.1(-) T cells showed an experienced/activated (CD44(HI)CD69(HI)CD122(HI)) cell phenotype, with high expression of Fas and PD-L1 correlating with their low proliferative capacity. Moreover, whereas IFN-gamma production by CD4(+)NK1.1(+) cells peaked at day 4 p.i., the IFN-gamma response of CD4(+)NK1.1(-) cells continued to increase at day 5 of infection. We also observed, at day 7 p.i., 2-fold higher percentages of perforin(+) cells in CD8(+)NK1.1(+) cells compared to CD8(+)NK1.1(-) cells. These results indicate that spleen NK1.1(+) and NK1.1(-) T cells respond to acute P. chabaudi malaria with different kinetics in terms of activation, proliferation, and IFN-gamma production.
Collapse
Affiliation(s)
- Sandra Marcia Muxel
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Chen J, Xu W, Zhou T, Ding Y, Duan J, Huang F. Inhibitory role of toll-like receptors agonists in Plasmodium yoelii liver stage development. Parasite Immunol 2009; 31:466-73. [PMID: 19646211 DOI: 10.1111/j.1365-3024.2009.01126.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
It is well known that innate immune plays an important role in controlling the development of Plasmodium liver stage. However, little is known about the role of toll-like receptors (TLR) signalling in the pre-erythrocytic immunity against Plasmodium. Here, we found that pre-treatment with individual TLR agonist pam3CSK4 (TLR2), poly(I:C) (TLR3), LPS (TLR4) and CpG (TLR9) could decrease significantly the liver malaria parasite load in mice for 58%, 63%, 75% and 88% respectively. Moreover, no parasitaemia was observed within 14 days in CpG group mice challenged with 100 sporozoites. At 24 h prior to CpG injection, administration of gadolinium chloride (GdCl(3)) led to the rebound of liver Plasmodium load through inhibiting selectively Kupffer cells (KC) phagocytosis capacity but failed to neutralize completely CpG-induced immunity against malaria liver stage. Compared with the control, pre-treatment of CpG up-regulated hepatic pro-inflammatory cytokines IL-12, IFN-gamma and TNF-alpha, but down-regulated anti-inflammatory cytokines IL-10 and TGF-beta. Hence, our data demonstrated the inhibitory role of diverse TLR agonists in the Plasmodium development during pre-erythrocytic stage. As the most robust agonist, CpG might inhibit the development of Plasmodium liver stage through regulation of intrahepatic inflammatory cytokines and enhancement of KC cells phagocytosis capacity.
Collapse
Affiliation(s)
- J Chen
- Department of Pathogenic Biology, Third Military Medical University, Chongqing 400038 China
| | | | | | | | | | | |
Collapse
|
23
|
A double-edged sword: the role of NKT cells in malaria and HIV infection and immunity. Semin Immunol 2009; 22:87-96. [PMID: 19962909 DOI: 10.1016/j.smim.2009.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 11/02/2009] [Accepted: 11/09/2009] [Indexed: 02/08/2023]
Abstract
NKT cells are known to play a role against certain microbial infections, including malaria and HIV, two major global infectious diseases. NKT cells exhibit either protective or pathogenic role against malaria. They are depleted by HIV infection and have a direct pathogenic role against many opportunistic infections common in end-stage AIDS. This review discusses the various features of the interaction between NKT cells and malaria parasites and HIV, and the potential to harness this interaction for therapeutic and vaccine strategies.
Collapse
|
24
|
Gameiro J, Nagib PRA, Andrade CF, Villa-Verde DMS, Silva-Barbosa SD, Savino W, Costa FTM, Verinaud L. Changes in cell migration-related molecules expressed by thymic microenvironment during experimental Plasmodium berghei infection: consequences on thymocyte development. Immunology 2009; 129:248-56. [PMID: 19824923 DOI: 10.1111/j.1365-2567.2009.03177.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We previously showed alterations in the thymus during experimental infection with Plasmodium berghei. Such alterations comprised histological changes, with loss of cortical-medullary limits, and the intrathymic presence of parasites. As the combination of chemokines, adhesion molecules and extracellular matrix (ECM) is critical to appropriate thymocyte development, we analysed the thymic expression of ECM ligands and receptors, as well as chemokines and their respective receptors during the experimental P. berghei infection. Increased expression of ECM components was observed in thymi from infected mice. In contrast, down-regulated surface expression of fibronectin and laminin receptors was observed in thymocytes from these animals. Moreover, in thymi from infected mice there was increased CXCL12 and CXCR4, and a decreased expression of CCL25 and CCR9. An altered thymocyte migration towards ECM elements and chemokines was seen when the thymi from infected mice were analysed. Evaluation of ex vivo migration patterns of CD4/CD8-defined thymocyte subpopulations revealed that double-negative (DN), and CD4(+) and CD8(+) single-positive (SP) cells from P. berghei-infected mice have higher migratory responses compared with controls. Interestingly, increased numbers of DN and SP subpopulations were found in the spleens of infected mice. Overall, we show that the thymic atrophy observed in P. berghei-infected mice is accompanied by thymic microenvironmental changes that comprise altered expression of thymocyte migration-related molecules of the ECM and chemokine protein families, which in turn can alter the thymocyte migration pattern. These thymic disturbances may have consequences for the control of the immune response against this protozoan.
Collapse
Affiliation(s)
- Jacy Gameiro
- Department of Anatomy, Cell Biology and Physiology, Institute of Biology, State University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Soulard V, Roland J, Gorgette O, Barbier E, Cazenave PA, Pied S. An early burst of IFN-gamma induced by the pre-erythrocytic stage favours Plasmodium yoelii parasitaemia in B6 mice. Malar J 2009; 8:128. [PMID: 19508725 PMCID: PMC2699347 DOI: 10.1186/1475-2875-8-128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Accepted: 06/09/2009] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND In murine models of malaria, an early proinflammatory response has been associated with the resolution of blood-stage infection. To dissect the protective immune mechanism that allow the control of parasitaemia, the early immune response of C57BL/6 mice induced during a non-lethal plasmodial infection was analysed. METHODS Mice were infected with Plasmodium yoelii 265BY sporozoites, the natural invasive form of the parasite, in order to complete its full life cycle. The concentrations of three proinflammatory cytokines in the sera of mice were determined by ELISA at different time points of infection. The contribution of the liver and the spleen to this cytokinic response was evaluated and the cytokine-producing lymphocytes were identified by flow cytometry. The physiological relevance of these results was tested by monitoring parasitaemia in genetically deficient C57BL/6 mice or wild-type mice treated with anti-cytokine neutralizing antibody. Finally, the cytokinic response in sera of mice infected with parasitized-RBCs was analysed. RESULTS The early immune response of C57BL/6 mice to sporozoite-induced malaria is characterized by a peak of IFN-gamma in the serum at day 5 of infection and splenic CD4 T lymphocytes are the major producer of this cytokine at this time point. Somewhat unexpected, the parasitaemia is significantly lower in P. yoelii-infected mice in the absence of IFN-gamma. More precisely, at early time points of infection, IFN-gamma favours parasitaemia, whereas helping to clear efficiently the blood-stage parasites at later time points. Interestingly, the early IFN-gamma burst is induced by the pre-erythrocytic stage. CONCLUSION These results challenge the current view regarding the role of IFN-gamma on the control of parasite growth since they show that IFN-gamma is not an essential mediator of protection in P. yoelii-infected C57BL/6 mice. Moreover, the mice parasitaemia is more efficiently controlled in the absence of an early IFN-gamma production, suggesting that this cytokine promotes parasite's growth. Finally, this early burst of IFN-gamma is induced by the pre-erythrocytic stage, showing the impact of this stage on the immune response taking place during the subsequent erythrocytic stage.
Collapse
Affiliation(s)
- Valérie Soulard
- Unité d'Immunophysiopathologie Infectieuse, Centre National de la Recherche Scientifique (CNRS) URA 1961, Université Paris VI, Institut Pasteur, Paris, France.
| | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Experimental malaria infection triggers early expansion of natural killer cells. Infect Immun 2008; 76:5873-82. [PMID: 18824529 DOI: 10.1128/iai.00640-08] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In order to gain a better understanding of gene expression during early malaria infection, we conducted microarray analysis of early blood responses in mice infected with erythrocytic-stage Plasmodium chabaudi. Immediately following infection, we observed coordinated and sequential waves of immune responses, with interferon-associated gene transcripts dominating by 16 h postinfection, followed by strong increases in natural killer (NK) cell-associated and major histocompatibility complex class I-related transcripts by 32 h postinfection. We showed by flow cytometry that the observed elevation in NK cell-associated transcripts was the result of a dramatic increase in the proportion of NK cells in the blood during infection. Subsequent microarray analysis of NK cells isolated from the peripheral blood of infected mice revealed a cell proliferation expression signature consistent with the observation that NK cells replicate in response to infection. Early proliferation of NK cells was directly observed in studies with adoptively transferred cells in infected mice. These data indicate that the early response to P. chabaudi infection of the blood is marked by a primary wave of interferon with a subsequent response by NK cells.
Collapse
|