1
|
Peng X, Chinwe Oluchi-Amaka I, Kwak-Kim J, Yang X. A comprehensive review of the roles of T-cell immunity in preeclampsia. Front Immunol 2025; 16:1476123. [PMID: 39981257 PMCID: PMC11841438 DOI: 10.3389/fimmu.2025.1476123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
Preeclampsia (PE) is an obstetrical disorder that occurs after the 20th week of gestation. It is recognized as one of the "Great Obstetrical Syndromes" and principally contributes to maternal morbidity and mortality. PE has been associated with a range of immune disorders, including a preponderance of T helper (Th) 1 over Th2 cells and imbalanced levels of Th17 and T regulatory cells (Tregs). During pregnancy, T cells safeguard the placenta against immune rejection and aid embryo implantation while involved in pregnancy complications, such as PE. Promoting alloantigen-specific Treg cells is a potential preventive and therapeutic strategy for PE. However, ensuring the safety of mothers and infants is of the utmost importance since the risk-benefit ratio of reproductive and obstetric conditions differs significantly from that of immune diseases that pose a life-threatening risk. In this review, we systematically summarize the roles of T-cell immunity in the peripheral blood, reproductive tissues, and at the maternal-fetal interface of PE patients. Furthermore, the recent therapeutic approaches centered on targeting T cell immunity in PE are critically appraised.
Collapse
Affiliation(s)
- Xu Peng
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| | | | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Clinical Immunology Laboratory, Foundational Sciences and Humanities, Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Xiuhua Yang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Sossou D, Ezinmegnon S, Agbota G, Gbedande K, Accrombessi M, Massougbodji A, d’Almeida M, Alao JM, Dossou-Dagba I, Pachot A, Vachot L, Brengel-Pesce K, Cottrell G, Yessoufou A, Briand V, Tissières P, Fievet N. Regulatory T cell homing and activation is a signature of neonatal sepsis. Front Immunol 2024; 15:1420554. [PMID: 39072327 PMCID: PMC11272980 DOI: 10.3389/fimmu.2024.1420554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/13/2024] [Indexed: 07/30/2024] Open
Abstract
Regulatory T cells (Treg) play a prominent role in utero tolerating non-inherited maternal antigens and in regulating immune responses against pathogens at birth. This study investigates Treg immunity in newborns in West Africa, where sepsis remains a major public health problem. Treg phenotypes on neonates subgroups with early-onset sepsis (EOS), presumed sepsis, and healthy newborn with and without prenatal risk factors were evaluated. Treg phenotypes varied according to prenatal conditions, with increase in Treg frequency and Foxp3 expression in healthy newborns with prenatal risk factors compared to those with none risk. Compared to healthy newborns with prenatal risk factors, EOS neonates had a significantly reduced frequency of Treg and Foxp3 expression. In the Treg pool, higher frequency of activated Treg was observed in EOS neonates, suggesting an in-utero activation upstream of the sepsis onset. Their migration to the infection site may explain the reduced frequency of circulating Integrin α4β1+ Treg suggestive of homing to the endothelial tissue. EOS neonates show increases expression of CTLA-4, PD-1 and CD39 on Treg, which negatively regulate the activation of effector T cells (Teff) corroborating by the lower frequency of Teff in EOS neonates. The higher frequency of CD39+ Treg and the lower frequency of integrinα4β1+ Treg in EOS non-survivor suggests that Treg exhaustement and endothelial homing are associated with outcome severity. Neonates developing EOS are born with an altered Treg phenotypic profile. Treg expression of CTLA-4, PD-1, CD39, and integrinα4β1 cell markers can be considered as early warning or diagnostic markers of EOS.
Collapse
Affiliation(s)
- Darius Sossou
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
- Faculty of Sciences and Technology (FAST), University of Abomey-Calavi, Institute of Applied Biomedical Sciences (ISBA), Laboratory of Cell Biology and Physiology, Cotonou, Benin
- Institut de Recherche Clinique du Bénin (IRCB), Calavi, Benin
| | - Sem Ezinmegnon
- Faculty of Sciences and Technology (FAST), University of Abomey-Calavi, Institute of Applied Biomedical Sciences (ISBA), Laboratory of Cell Biology and Physiology, Cotonou, Benin
- Fédérations Hospitalo-Universitaires (FHU) Sepsis, AP-HP/Université Paris Saclay/Inserm, Le Kremlin-Bicêtre, France
| | - Gino Agbota
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Calavi, Benin
| | - Komi Gbedande
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Calavi, Benin
| | - Manfred Accrombessi
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Calavi, Benin
| | | | - Marceline d’Almeida
- Pediatric Department, National University Hospital Center (CNHU), Cotonou, Benin
| | - Jules M. Alao
- Pediatric Department, Mother and Child University and Hospital Center (CHUMEL), Cotonou, Benin
| | | | - Alexandre Pachot
- Medical Diagnostic Discovery Department, bioMérieux, Marcy l’Etoile, France
| | - Laurence Vachot
- Medical Diagnostic Discovery Department, bioMérieux, Marcy l’Etoile, France
| | | | - Gilles Cottrell
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
| | - Akadiri Yessoufou
- Faculty of Sciences and Technology (FAST), University of Abomey-Calavi, Institute of Applied Biomedical Sciences (ISBA), Laboratory of Cell Biology and Physiology, Cotonou, Benin
| | - Valérie Briand
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
| | - Pierre Tissières
- Fédérations Hospitalo-Universitaires (FHU) Sepsis, AP-HP/Université Paris Saclay/Inserm, Le Kremlin-Bicêtre, France
- Institute of Integrative Biology of the Cell (I2BC), Centre National de la Recherche Scientifique (CNRS), Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), University Paris Saclay, Gif-sur-Yvette, France
- Pediatric Intensive Care and Neonatal Medicine, Assistance Publique - Hôpitaux de Paris (AP-HP) Paris Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Nadine Fievet
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
| |
Collapse
|
3
|
Abdulla ZA, Al-Bashir SM, Alzoubi H, Al-Salih NS, Aldamen AA, Abdulazeez AZ. The Role of Immunity in the Pathogenesis of SARS-CoV-2 Infection and in the Protection Generated by COVID-19 Vaccines in Different Age Groups. Pathogens 2023; 12:329. [PMID: 36839601 PMCID: PMC9967364 DOI: 10.3390/pathogens12020329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
This study aims to review the available data regarding the central role of immunity in combating SARS-CoV-2 infection and in the generation of protection by vaccination against COVID-19 in different age groups. Physiologically, the immune response and the components involved in it are variable, both functionally and quantitatively, in neonates, infants, children, adolescents, and adults. These immunological differences are mirrored during COVID-19 infection and in the post-vaccination period. The outcome of SARS-CoV-2 infection is greatly dependent on the reaction orchestrated by the immune system. This is clearly obvious in relation to the clinical status of COVID-19 infection, which can be symptomless, mild, moderate, or severe. Even the complications of the disease show a proportional pattern in relation to the immune response. On the contrary, the commonly used anti-COVID-19 vaccines generate protective humoral and cellular immunity. The magnitude of this immunity and the components involved in it are discussed in detail. Furthermore, many of the adverse effects of these vaccines can be explained on the basis of immune reactions against the different components of the vaccines. Regarding the appropriate choice of vaccine for different age groups, many factors have to be considered. This is a cornerstone, particularly in the following age groups: 1 day to 5 years, 6 to 11 years, and 12 to 17 years. Many factors are involved in deciding the route, doses, and schedule of vaccination for children. Another important issue in this dilemma is the hesitancy of families in making the decision about whether to vaccinate their children. Added to these difficulties is the choice by health authorities and governments concerning whether to make children's vaccination compulsory. In this respect, although rare and limited, adverse effects of vaccines in children have been detected, some of which, unfortunately, have been serious or even fatal. However, to achieve comprehensive control over COVID-19 in communities, both children and adults have to be vaccinated, as the former group represents a reservoir for viral transmission. The understanding of the various immunological mechanisms involved in SARS-CoV-2 infection and in the preparation and application of its vaccines has given the sciences a great opportunity to further deepen and expand immunological knowledge. This will hopefully be reflected positively on other diseases through gaining an immunological background that may aid in diagnosis and therapy. Humanity is still in continuous conflict with SARS-CoV-2 infection and will be for a while, but the future is expected to be in favor of the prevention and control of this disease.
Collapse
Affiliation(s)
| | - Sharaf M. Al-Bashir
- Department of Clinical Sciences, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan
| | - Hiba Alzoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan
| | - Noor S. Al-Salih
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan
| | - Ala A. Aldamen
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan
| | | |
Collapse
|
4
|
Kalia I, Anand R, Quadiri A, Bhattacharya S, Sahoo B, Singh AP. Plasmodium berghei-Released Factor, PbTIP, Modulates the Host Innate Immune Responses. Front Immunol 2022; 12:699887. [PMID: 34987497 PMCID: PMC8721568 DOI: 10.3389/fimmu.2021.699887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
The Plasmodium parasite has to cross various immunological barriers for successful infection. Parasites have evolved mechanisms to evade host immune responses, which hugely contributes to the successful infection and transmission by parasites. One way in which a parasite evades immune surveillance is by expressing molecular mimics of the host molecules in order to manipulate the host responses. In this study, we report a Plasmodium berghei hypothetical protein, PbTIP (PbANKA_124360.0), which is a Plasmodium homolog of the human T-cell immunomodulatory protein (TIP). The latter possesses immunomodulatory activities and suppressed the host immune responses in a mouse acute graft-versus-host disease (GvHD) model. The Plasmodium berghei protein, PbTIP, is expressed on the merozoite surface and exported to the host erythrocyte surface upon infection. It is shed in the blood circulation by the activity of an uncharacterized membrane protease(s). The shed PbTIP could be detected in the host serum during infection. Our results demonstrate that the shed PbTIP exhibits binding on the surface of macrophages and reduces their inflammatory cytokine response while upregulating the anti-inflammatory cytokines such as TGF-β and IL-10. Such manipulated immune responses are observed in the later stage of malaria infection. PbTIP induced Th2-type gene transcript changes in macrophages, hinting toward its potential to regulate the host immune responses against the parasite. Therefore, this study highlights the role of a Plasmodium-released protein, PbTIP, in immune evasion using macrophages, which may represent the critical strategy of the parasite to successfully survive and thrive in its host. This study also indicates the human malaria parasite TIP as a potential diagnostic molecule that could be exploited in lateral flow-based immunochromatographic tests for malaria disease diagnosis.
Collapse
Affiliation(s)
- Inderjeet Kalia
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| | - Rajesh Anand
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| | - Afshana Quadiri
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| | - Shreya Bhattacharya
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| | - Bijayalaxmi Sahoo
- Department of Biological Sciences and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| | - Agam Prasad Singh
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
5
|
Feyaerts D, Urbschat C, Gaudillière B, Stelzer IA. Establishment of tissue-resident immune populations in the fetus. Semin Immunopathol 2022; 44:747-766. [PMID: 35508672 PMCID: PMC9067556 DOI: 10.1007/s00281-022-00931-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/17/2022] [Indexed: 12/15/2022]
Abstract
The immune system establishes during the prenatal period from distinct waves of stem and progenitor cells and continuously adapts to the needs and challenges of early postnatal and adult life. Fetal immune development not only lays the foundation for postnatal immunity but establishes functional populations of tissue-resident immune cells that are instrumental for fetal immune responses amidst organ growth and maturation. This review aims to discuss current knowledge about the development and function of tissue-resident immune populations during fetal life, focusing on the brain, lung, and gastrointestinal tract as sites with distinct developmental trajectories. While recent progress using system-level approaches has shed light on the fetal immune landscape, further work is required to describe precise roles of prenatal immune populations and their migration and adaptation to respective organ environments. Defining points of prenatal susceptibility to environmental challenges will support the search for potential therapeutic targets to positively impact postnatal health.
Collapse
Affiliation(s)
- Dorien Feyaerts
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA
| | - Christopher Urbschat
- grid.13648.380000 0001 2180 3484Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | - Brice Gaudillière
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA USA
| | - Ina A. Stelzer
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA
| |
Collapse
|
6
|
Fievet N, Ezinmegnon S, Agbota G, Sossou D, Ladekpo R, Gbedande K, Briand V, Cottrell G, Vachot L, Yugueros Marcos J, Pachot A, Textoris J, Blein S, Lausten-Thomsen U, Massougbodji A, Bagnan L, Tchiakpe N, d'Almeida M, Alao J, Dossou-Dagba I, Tissieres P. SEPSIS project: a protocol for studying biomarkers of neonatal sepsis and immune responses of infants in a malaria-endemic region. BMJ Open 2020; 10:e036905. [PMID: 32709653 PMCID: PMC7380952 DOI: 10.1136/bmjopen-2020-036905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Neonatal sepsis outreaches all causes of neonatal mortality worldwide and remains a major societal burden in low and middle income countries. In addition to limited resources, endemic morbidities, such as malaria and prematurity, predispose neonates and infants to invasive infection by altering neonatal immune response to pathogens. Nevertheless, thoughtful epidemiological, diagnostic and immunological evaluation of neonatal sepsis and the impact of gestational malaria have never been performed. METHODS AND ANALYSIS A prospective longitudinal multicentre follow-up of 580 infants from birth to 3 months of age in urban and suburban Benin will be performed. At delivery, and every other week, all children will be examined and clinically evaluated for occurrence of sepsis. At delivery, cord blood systematic analysis of selected plasma and transcriptomic biomarkers (procalcitonin, interleukin (IL)-6, IL-10, IP10, CD74 and CX3CR1) associated with sepsis pathophysiology will be evaluated in all live births as well as during the follow-up, and when sepsis will be suspected. In addition, whole blood response to selected innate stimuli and extensive peripheral blood mononuclear cells phenotypic characterisation will be performed. Reference intervals specific to sub-Saharan neonates will be determined from this cohort and biomarkers performances for neonatal sepsis diagnosis and prognosis tested. ETHICS AND DISSEMINATION Ethical approval has been obtained from the Comité d'Ethique de la Recherche - Institut des Sciences Biomédicales Appliquées (CER-ISBA 85 - 5 April 2016, extended on 3 February 2017). Results will be disseminated through international presentations at scientific meetings and publications in peer-reviewed journals. TRIAL REGISTRATION NUMBER ClinicalTrials.gov registration number: NCT03780712.
Collapse
Affiliation(s)
- Nadine Fievet
- Institut de Recherche pour le Développement (IRD), Mère et enfant face aux infections tropicales (UMR216), Paris, France
- COMUE Sorbonne Paris Cité, Universite Paris Descartes, Paris, Île-de-France, France
| | - Sem Ezinmegnon
- Department of Microbiology, Institut de Biologie Integrative de la Cellule, Gif-sur-Yvette, France
- Medical Diagnostic Discovery Department (MD3), bioMerieux SA, Marcy l'Etoile, Rhône-Alpes, France
| | - Gino Agbota
- UMR216-MERIT, French National Research Institute for Sustainable Development (IRD), Université Paris Descartes, Paris, France
- Institut de Recherche Clinique du Bénin, Calavi, Benin
| | - Darius Sossou
- Institut de Recherche Clinique du Bénin, Calavi, Benin
| | | | - Komi Gbedande
- Institut de Recherche Clinique du Benin, Cotonou, Benin
| | - Valerie Briand
- Institut de Recherche pour le Développement (IRD), Mère et enfant face aux infections tropicales (UMR216), Paris, France
| | - Gilles Cottrell
- UMR216, Institut de Recherche pour le Développement, Cotonou, Benin
- Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Laurence Vachot
- Medical Diagnostic Discovery Department (MD3), bioMerieux SA, Marcy l'Etoile, Rhône-Alpes, France
| | - Javier Yugueros Marcos
- Medical Diagnostic Discovery Department (MD3), bioMerieux SA, Marcy l'Etoile, Rhône-Alpes, France
| | - Alexandre Pachot
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression, bioMerieux, LYON cedex 03, France
| | - Julien Textoris
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression, bioMerieux, LYON cedex 03, France
- Département d'Anesthésie et de Réanimation, Hospices Civils de Lyon, LYON Cedex 03, France
| | - Sophie Blein
- Medical Diagnostic Discovery Department (MD3), bioMerieux SA, Marcy l'Etoile, Rhône-Alpes, France
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression, bioMerieux, LYON cedex 03, France
| | - Ulrik Lausten-Thomsen
- Pediatric Intensive Care, Hopitaux Universitaires Paris-Sud, Le Kremlin-Bicetre, France
| | | | - Lehila Bagnan
- Institut de Recherche Clinique du Bénin, Calavi, Benin
- Department of Paediatric, National University Hospital Center (CNHU), Cotonou, Benin
| | - Nicole Tchiakpe
- Institut de Recherche Clinique du Bénin, Calavi, Benin
- Department of Paediatric, Centre Hospitalier Universitaire de la Mère et de l'Enfant Lagune (CHUMEL), Cotonou, Benin
| | - Marceline d'Almeida
- Department of Paediatric, National University Hospital Center (CNHU), Cotonou, Benin
- Institut de Recherche Clinique du Benin, Calavi, Île-de-France, Benin
| | | | | | - Pierre Tissieres
- Department of Microbiology, Institut de Biologie Integrative de la Cellule, Gif-sur-Yvette, France
- Pediatric Intensive Care, Hopitaux Universitaires Paris-Sud, Le Kremlin-Bicetre, France
| |
Collapse
|
7
|
Odorizzi PM, Jagannathan P, McIntyre TI, Budker R, Prahl M, Auma A, Burt TD, Nankya F, Nalubega M, Sikyomu E, Musinguzi K, Naluwu K, Kakuru A, Dorsey G, Kamya MR, Feeney ME. In utero priming of highly functional effector T cell responses to human malaria. Sci Transl Med 2019; 10:10/463/eaat6176. [PMID: 30333241 DOI: 10.1126/scitranslmed.aat6176] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/10/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022]
Abstract
Malaria remains a significant cause of morbidity and mortality worldwide, particularly in infants and children. Some studies have reported that exposure to malaria antigens in utero results in the development of tolerance, which could contribute to poor immunity to malaria in early life. However, the effector T cell response to pathogen-derived antigens encountered in utero, including malaria, has not been well characterized. Here, we assessed the frequency, phenotype, and function of cord blood T cells from Ugandan infants born to mothers with and without placental malaria. We found that infants born to mothers with active placental malaria had elevated frequencies of proliferating effector memory fetal CD4+ T cells and higher frequencies of CD4+ and CD8+ T cells that produced inflammatory cytokines. Fetal CD4+ and CD8+ T cells from placental malaria-exposed infants exhibited greater in vitro proliferation to malaria antigens. Malaria-specific CD4+ T cell proliferation correlated with prospective protection from malaria during childhood. These data demonstrate that placental malaria is associated with the generation of proinflammatory malaria-responsive fetal T cells. These findings add to our current understanding of fetal immunity and indicate that a functional and protective pathogen-specific T cell response can be generated in utero.
Collapse
Affiliation(s)
- Pamela M Odorizzi
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA
| | | | - Tara I McIntyre
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA
| | - Rachel Budker
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA
| | - Mary Prahl
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Ann Auma
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Trevor D Burt
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | - Esther Sikyomu
- Infectious Disease Research Collaboration, Kampala, Uganda
| | | | - Kate Naluwu
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Abel Kakuru
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA
| | - Moses R Kamya
- School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Margaret E Feeney
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110 USA. .,Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
8
|
Feeney ME. The immune response to malaria in utero. Immunol Rev 2019; 293:216-229. [PMID: 31553066 DOI: 10.1111/imr.12806] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022]
Abstract
Malaria causes tremendous early childhood morbidity and mortality, providing an urgent impetus for the development of a vaccine that is effective in neonates. However, the infant immune response to malaria may be influenced by events that occur well before birth. Placental malaria infection complicates one quarter of all pregnancies in Africa and frequently results in exposure of the fetus to malaria antigens in utero, while the immune system is still developing. Some data suggest that in utero exposure to malaria may induce immunologic tolerance that interferes with the development of protective immunity during childhood. More recently, however, a growing body of evidence suggests that fetal malaria exposure can prime highly functional malaria-specific T- and B-cells, which may contribute to postnatal protection from malaria. In utero exposure to malaria also impacts the activation and maturation of fetal antigen presenting cells and innate lymphocytes, which could have implications for global immunity in the infant. Here, we review recent advances in our understanding of how various components of the fetal immune system are altered by in utero exposure to malaria, discuss factors that may tilt the critical balance between tolerance and adaptive immunity, and consider the implications of these findings for malaria prevention strategies.
Collapse
Affiliation(s)
- Margaret E Feeney
- Departments of Pediatrics and Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Alam I, Almajwal AM, Alam W, Alam I, Ullah N, Abulmeaaty M, Razak S, Khan S, Pawelec G, Paracha PI. The immune-nutrition interplay in aging – facts and controversies. ACTA ACUST UNITED AC 2019. [DOI: 10.3233/nha-170034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Iftikhar Alam
- Department of Community Health Sciences, Clinical Nutrition Program, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
- Department of Human Nutrition & Dietetics, Bacha Khan University Charsadda, Charsadda, Khyber Pakhtunkhwa, Pakistan
- Tübingen Ageing and Tumour Immunology Group, Zentrum für Medizinische Forschung, University of Tübingen, Tübingen, Germany
| | - Ali M. Almajwal
- Department of Community Health Sciences, Clinical Nutrition Program, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Wajid Alam
- Oral and Maxillofacial Surgery, Khyber Colleg of Dentistry, KPK, Peshawar, Pakistan
| | - Ibrar Alam
- Department of Biotechnology, Bacha Khan University Charsadda, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Niamat Ullah
- Department of Human Nutrition, The Agriculture University Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Mahmoud Abulmeaaty
- Department of Community Health Sciences, Clinical Nutrition Program, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Razak
- Department of Community Health Sciences, Clinical Nutrition Program, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Saleem Khan
- Department of Human Nutrition, The Agriculture University Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Graham Pawelec
- Tübingen Ageing and Tumour Immunology Group, Zentrum für Medizinische Forschung, University of Tübingen, Tübingen, Germany
- Health Sciences North Research Institute, Sudbury, ON, Canada
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Parvez Iqbal Paracha
- Department of Human Nutrition, The Agriculture University Peshawar, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
10
|
Malhotra I, LaBeaud AD, Morris N, McKibben M, Mungai P, Muchiri E, King CL, King CH. Cord Blood Antiparasite Interleukin 10 as a Risk Marker for Compromised Vaccine Immunogenicity in Early Childhood. J Infect Dis 2019; 217:1426-1434. [PMID: 29390149 DOI: 10.1093/infdis/jiy047] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 01/23/2018] [Indexed: 01/09/2023] Open
Abstract
Background Antenatal exposure to parasites can affect infants' subsequent responses to vaccination. The present study investigated how maternal prenatal infections and newborns' antiparasite cytokine profiles relate to immunoglobulin G (IgG) responses to standard vaccination during infancy. Methods A total of 450 Kenyan women were tested for parasitic infections during pregnancy. Their newborns' responses to Plasmodium falciparum, schistosome, and filaria antigens were assessed in cord blood lymphocytes. Following standard neonatal vaccination, this infant cohort was followed biannually to age 30 months for measurement of circulating IgG levels against Haemophilus influenzae b (Hib), diphtheria toxoid (DT), hepatitis B virus (HBV), and tetanus toxoid. Results Trajectories of postvaccination IgG levels were classified by functional principal component (PC) analysis to assess each child's response profile. Two main components, PC1, reflecting height of response over time, and PC2, reflecting crossover from high to low responses or from low to high responses, were identified. Cord blood cytokine responses to schistosome and filarial antigens showed a significant association between augmented antihelminth interleukin 10 and reduced antibody levels, particularly to DT and HBV, and a more rapid postvaccination decline in circulating IgG levels against Hib. Conclusion Antenatal sensitization to schistosomiasis or filariasis and related production of antiparasite interleukin 10 at birth are associated with reduced antivaccine IgG levels in infancy, with possibly impaired protection.
Collapse
Affiliation(s)
- Indu Malhotra
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio.,Clinical and Translational Science Collaborative, Case Western Reserve University, Cleveland, Ohio
| | - A Desiree LaBeaud
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio.,Child Health Research Institute, Stanford, California
| | - Nathan Morris
- Clinical and Translational Science Collaborative, Case Western Reserve University, Cleveland, Ohio
| | - Maxim McKibben
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio
| | - Peter Mungai
- Division of Vector Borne and Neglected Tropical Diseases, Ministry of Public Health and Sanitation, Nairobi, Kenya
| | - Eric Muchiri
- Division of Vector Borne and Neglected Tropical Diseases, Ministry of Public Health and Sanitation, Nairobi, Kenya
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio
| | - Charles H King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
11
|
Harrington WE, Kakuru A, Jagannathan P. Malaria in pregnancy shapes the development of foetal and infant immunity. Parasite Immunol 2018; 41:e12573. [PMID: 30019470 DOI: 10.1111/pim.12573] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Malaria, particularly Plasmodium falciparum, continues to disproportionately affect pregnant women. In addition to the profoundly deleterious impact of maternal malaria on the health of the mother and foetus, malaria infection in pregnancy has been shown to affect the development of the foetal and infant immune system and may alter the risk of malaria and nonmalarial outcomes during infancy. This review summarizes our current understanding of how malaria infection in pregnancy shapes the protective components of the maternal immune system transferred to the foetus and how foetal exposure to parasite antigens impacts the development of foetal and infant immunity. It also reviews existing evidence linking malaria infection in pregnancy to malaria and nonmalarial outcomes in infancy and how preventing malaria in pregnancy may alter these outcomes. A better understanding of the consequences of malaria infection in pregnancy on the development of foetal and infant immunity will inform control strategies, including intermittent preventive treatment in pregnancy and vaccine development.
Collapse
Affiliation(s)
- Whitney E Harrington
- Department of Pediatrics, University of Washington/Seattle Children's Hospital, Seattle, Washington
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | |
Collapse
|
12
|
Interferon- γ and Interleukin-10 Responses during Clinical Malaria Episodes in Infants Aged 0-2 Years Prenatally Exposed to Plasmodium falciparum: Tanzanian Birth Cohort. J Trop Med 2018; 2018:6847498. [PMID: 30154871 PMCID: PMC6091450 DOI: 10.1155/2018/6847498] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 06/21/2018] [Accepted: 07/11/2018] [Indexed: 12/17/2022] Open
Abstract
Background Infants born to mothers with placental malaria are prenatally exposed to Plasmodium falciparum antigens. However, the effect of that exposure to subsequent immune responses has not been fully elucidated. This study aimed at determining the effect of prenatal exposure to P. falciparum on Interleukin-10 and Interferon-γ responses during clinical malaria episodes in the first 24 months of life. Methods This prospective cohort study involved 215 infants aged 0-2 years born to mothers with or without placental malaria. Enzyme-linked immunosorbent assay (ELISA) was used to determine levels of IL-10 and IFN-γ in infants and detect IgM in cord blood. Data were analyzed using SPSS version 20. Findings Geometric mean for IFN-γ in exposed infants was 557.9 pg/ml (95% CI: 511.6-604.1) and in unexposed infants it was 634.4 pg/ml (95% CI: 618.2-668.5) (P=0.02). Mean IL-10 was 22.4 pg/ml (95% CI: 19.4-28.4) and 15.1 pg/ml (95%CI: 12.4-17.6), respectively (P=0.01). Conclusions Prenatal exposure to P. falciparum antigens significantly affects IL-10 and IFN-γ responses during clinical malaria episodes in the first two years of life.
Collapse
|
13
|
Chen L, Bennett E, Wheeler AJ, Lyons AB, Woods GM, Johnston F, Zosky GR. Maternal exposure to particulate matter alters early post-natal lung function and immune cell development. ENVIRONMENTAL RESEARCH 2018; 164:625-635. [PMID: 29627759 DOI: 10.1016/j.envres.2018.03.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND In utero exposure to particulate matter (PM) from a range of sources is associated with adverse post-natal health; however, the effect of maternal exposure to community-sampled PM on early post-natal lung and immune development is poorly understood. OBJECTIVES Using a mouse model, we aimed to determine whether in utero exposure to PM alters early post-natal lung function and immune cell populations. We used PM collected from ceiling voids in suburban houses as a proxy for community PM exposure. METHODS Pregnant C57BL/6 mice were intranasally exposed to ceiling derived PM, or saline alone, at gestational day (E) 13.5, 15.5, and 17.5. When mice were two weeks old, we assessed lung function by the forced oscillation technique, and enumerated T and B cell populations in the spleen and thymus by flow cytometry. RESULTS Maternal exposure to PM impaired somatic growth of male offspring resulting in reduced lung volume and deficits in lung function. There was no effect on thymic T cell populations in dams and their male offspring but PM decreased the CD4 +CD25 + T cell population in the female offspring. In contrast, maternal exposure to PM increased splenic CD3 +CD4 + and CD3 +CD8 + T cells in dams, and there was some evidence to suggest inhibition of splenic T cell maturation in male but not female offspring. CONCLUSIONS Our findings suggested that maternal exposure to ceiling void PM has the capacity to impair early somatic growth and alter early life immune development in a sex specific manner.
Collapse
Affiliation(s)
- Ling Chen
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2308, Australia; School of Medicine, Faculty of Health, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Ellen Bennett
- School of Medicine, Faculty of Health, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Amanda J Wheeler
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - A Bruce Lyons
- School of Medicine, Faculty of Health, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Gregory M Woods
- Cancer and Immunology Research Group, Menzies Research Institute, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Fay Johnston
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Graeme R Zosky
- School of Medicine, Faculty of Health, University of Tasmania, Hobart, Tasmania 7000, Australia.
| |
Collapse
|
14
|
Sylvester B, Gasarasi DB, Aboud S, Tarimo D, Massawe S, Mpembeni R, Swedberg G. Hyperparasitaemia during clinical malaria episodes in infants aged 0-24 months and its association with in utero exposure to Plasmodium falciparum. BMC Res Notes 2018; 11:232. [PMID: 29618382 PMCID: PMC5885461 DOI: 10.1186/s13104-018-3339-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 03/30/2018] [Indexed: 11/18/2022] Open
Abstract
Objective Existing information has shown that infants who are prenatally exposed to P. falciparum are susceptible to subsequent malaria infections. However, the effect of prenatal exposure to P. falciparum on parasite density during clinical malaria episodes has not been fully elucidated. This study is a component of a prospective cohort study for which initial results have been published. This component was designed to determine the effect of prenatal exposure to P. falciparum on parasite density during clinical malaria episodes in the first 24 months of life. A total of 215 infants were involved and monitored for clinical malaria episodes defined by fever (≥ 37 °C) and parasitaemia. The geometric mean parasite counts between exposed and unexposed infants were compared using independent samples t test. The effect of in utero exposure to P. falciparum on parasite density was assessed using binary logistic regression. Results The geometric mean parasite count per µl of blood during clinical malaria episodes in exposed infants was 24,889 (95% CI 18,286–31,490) while in unexposed infants it was 14,035 (95% CI 12,111–15,960), P < 0.05. Prenatal exposure to P. falciparum was associated with hyperparasitaemia during clinical malaria episodes (OR 7.04, 95% CI 2.31–21.74), while other factors were not significantly associated (P > 0.05).
Collapse
Affiliation(s)
- Boniphace Sylvester
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania.
| | - Dinah B Gasarasi
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Donath Tarimo
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Siriel Massawe
- Department of Obstetrics and Gynaecology, School of Medicine, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Rose Mpembeni
- Department of Epidemiology and Biostatistics, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Gote Swedberg
- Department of Medical Biochemistry and Microbiology, Biomedical Centre, Uppsala University, P.O.BOX 582, Uppsala, Sweden
| |
Collapse
|
15
|
Nguyen TA, Kahn DA, Loewendorf AI. Placental implantation over prior cesarean scar causes activation of fetal regulatory T cells. IMMUNITY INFLAMMATION AND DISEASE 2018; 6:256-263. [PMID: 29430878 PMCID: PMC5946153 DOI: 10.1002/iid3.214] [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: 10/09/2017] [Revised: 12/18/2017] [Accepted: 01/05/2018] [Indexed: 11/10/2022]
Abstract
Introduction Maternal‐fetal chimerism is miniscule, a testament to the integrity of the uteroplacental interface. The soundness of this border region is potentially altered through cesarean delivery of prior babies with uncertain consequences for the following pregnancies. Methods Using multicolor flow cytometry and quantitative PCR of non‐inherited maternal antigens we performed a retrospective case control pilot study and formulated the null hypothesis that placental implantation over a prior uterine scar does not result in the presence of memory Treg (CD45RO+) in the fetus. We then performed a power calculation and performed a blinded, appropriately powered prospective case control study to test the null hypothesis. Results Fetuses born to mothers with prior uterine scar have a roughly five times higher maternal to fetal microchimerism when the placenta directly interacts with the uterine scar. Unlike exposure to antigens in adult life, in utero antigenic exposure induces tolerogenic (Treg) responses in fetuses and we here report the presence of fetal Treg with a memory phenotype (CD45RO+). However, we only find such CD45RO+ fetal Tregs when the placenta abuts the uterine scar (Risk Ratio = 5 [p < 0.05 CI:(1.448 to 17.27)]). These memory fetal Tregs are functionally highly suppressive compared to CD45RA‐expressing fetal Tregs, and have specificity for non‐inherited maternal antigens. Conclusions We found that uterine scars, in the case of our study these scars are from prior c‐sections, fundamentally impair uterine integrity allowing for increased antigen exposure of the fetus; with our appropriately powered study we rejected the null hypothesis and accepted the alternative hypothesis that placental implantation over a prior uterine scar results in the presence of memory Treg (CD45RO+) in the fetus. Thus, our study demonstrates a previously unappreciated role for uterine integrity in limiting fetal antigenic exposure, a key element to avoid the formation of inappropriate tolerances by the fundamentally tolerogenic fetal immune system.
Collapse
Affiliation(s)
- Tina A Nguyen
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine,UCLA, Los Angeles, California
| | - Daniel A Kahn
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine,UCLA, Los Angeles, California.,Asante Physician Partners, Medford, Oregon
| | - Andrea I Loewendorf
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine,UCLA, Los Angeles, California.,Huntington Medical Research Institutes, Reproductive - and Vascular Immunology, Pasadena, California
| |
Collapse
|
16
|
Tassi Yunga S, Fouda GG, Sama G, Ngu JB, Leke RGF, Taylor DW. Increased Susceptibility to Plasmodium falciparum in Infants is associated with Low, not High, Placental Malaria Parasitemia. Sci Rep 2018; 8:169. [PMID: 29317740 PMCID: PMC5760570 DOI: 10.1038/s41598-017-18574-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/12/2017] [Indexed: 01/19/2023] Open
Abstract
Risk of malaria in infants can be influenced by prenatal factors. In this study, the potential for placental parasitemia at delivery in predicting susceptibility of infants to Plasmodium falciparum (Pf) infections was evaluated. Seventy-two newborns of mothers who were placental malaria negative (PM-) and of mothers who were PM+ with below (PM+ Lo) and above (PM + Hi) median placental parasitemia, were actively monitored during their first year of life. Median time to first PCR-detected Pf infection was shorter in PM + Lo infants (2.8 months) than in both PM- infants (4.0 months, p = 0.002) and PM + Hi infants (4.1 months, p = 0.01). Total number of new infections was also highest in the PM + Lo group. Only 24% of infants experienced clinical malaria episodes but these episodes occurred earlier in PM + Lo infants than in PM + Hi infants (p = 0.05). The adjusted hazard ratio (95% CI) of having Pf infection was 3.9 (1.8-8.4) and 1.5 (0.7-3.4) for infants in the PM + Lo and PM + Hi groups, respectively. Collectively, low placental parasitemia was associated with increased susceptibility to malaria during infancy. Therefore, malaria in pregnancy preventive regimens, such as sulfadoxine-pyremethamine, that reduce but do not eliminate placental Pf in areas of drug resistance may increase the risk of malaria in infants.
Collapse
Affiliation(s)
- Samuel Tassi Yunga
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, BSB320, Honolulu, HI, 96813, USA
| | - Genevieve G Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA and Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Grace Sama
- The Biotechnology Center, University of Yaoundé 1, BP 3851 Messa, Yaoundé, Cameroon
| | - Julia B Ngu
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, BP 1364, Yaoundé, Cameroon
| | - Rose G F Leke
- The Biotechnology Center, University of Yaoundé 1, BP 3851 Messa, Yaoundé, Cameroon
| | - Diane W Taylor
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, BSB320, Honolulu, HI, 96813, USA.
| |
Collapse
|
17
|
El-Chennawi F, Rageh IM, Mansour AI, Darwish MI, Elghzaly AA, Sakr BES, Elbaz KM. Comparison of the percentages of CD4+
CD25high
FOXP3+
, CD4+
CD25low
FOXP3+
, and CD4+
FOXP3+
Tregs, in the umbilical cord blood of babies born to mothers with and without preeclampsia. Am J Reprod Immunol 2017; 78. [DOI: 10.1111/aji.12761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/21/2017] [Indexed: 01/03/2023] Open
Affiliation(s)
- Farha El-Chennawi
- Clinical and Chemical Pathology Department; Faculty of Medicine; Mansoura University; Mansoura Egypt
| | - Ibrahim Mohamed Rageh
- Clinical and Chemical Pathology Department; Faculty of Medicine; Benha University; Benha Egypt
| | - Amira Ibrahim Mansour
- Clinical and Chemical Pathology Department; Faculty of Medicine; Benha University; Benha Egypt
| | - Mohammed Ibrahim Darwish
- Clinical and Chemical Pathology Department; Faculty of Medicine; Mansoura University; Mansoura Egypt
| | - Ashraf Antar Elghzaly
- Clinical and Chemical Pathology Department; Faculty of Medicine; Mansoura University; Mansoura Egypt
| | - Basma El Sayed Sakr
- Gynecology and Obstetrics Department; Faculty of Medicine; Benha University; Benha Egypt
| | | |
Collapse
|
18
|
Harrington WE, Kanaan SB, Muehlenbachs A, Morrison R, Stevenson P, Fried M, Duffy PE, Nelson JL. Maternal Microchimerism Predicts Increased Infection but Decreased Disease due to Plasmodium falciparum During Early Childhood. J Infect Dis 2017; 215:1445-1451. [PMID: 28329160 DOI: 10.1093/infdis/jix129] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/08/2017] [Indexed: 11/13/2022] Open
Abstract
Background A mother's infection with placental malaria (PM) can affect her child's susceptibility to malaria, although the mechanism remains unclear. The fetus acquires a small amount of maternal cells and DNA known as maternal microchimerism (MMc), and we hypothesized that PM increases MMc and that MMc alters risk of Plasmodium falciparum malaria during infancy. Methods In a nested cohort from Muheza, Tanzania, we evaluated the presence and level of cord blood MMc in offspring of women with and without PM. A maternal-specific polymorphism was identified for each maternal-infant pair, and MMc was assayed by quantitative polymerase chain reaction. The ability of MMc to predict malaria outcomes during early childhood was evaluated in longitudinal models. Results Inflammatory PM increased the detection rate of MMc among offspring of primigravidae and secundigravidae, and both noninflammatory and inflammatory PM increased the level of MMc. Detectable MMc predicted increased risk of positive blood smear but, interestingly, decreased risk of symptomatic malaria and malaria hospitalization. Conclusions The acquisition of MMc may result in increased malaria infection but protection from malaria disease. Future studies should be directed at the cellular component of MMc, with attention to its ability to directly or indirectly coordinate anti-malarial immune responses in the offspring.
Collapse
Affiliation(s)
- Whitney E Harrington
- Department of Pediatrics, University of Washington School of Medicine/Seattle Children's Hospital, Washington
| | - Sami B Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Atis Muehlenbachs
- Department of Pathology, University of Washington, Seattle, Washington
| | - Robert Morrison
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Philip Stevenson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - J Lee Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Rheumatology, University of Washington, Seattle
| |
Collapse
|
19
|
Montes de Oca M, Good MF, McCarthy JS, Engwerda CR. The Impact of Established Immunoregulatory Networks on Vaccine Efficacy and the Development of Immunity to Malaria. THE JOURNAL OF IMMUNOLOGY 2016; 197:4518-4526. [DOI: 10.4049/jimmunol.1600619] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/26/2016] [Indexed: 02/07/2023]
|
20
|
Timing of in utero malaria exposure influences fetal CD4 T cell regulatory versus effector differentiation. Malar J 2016; 15:497. [PMID: 27717402 PMCID: PMC5055709 DOI: 10.1186/s12936-016-1545-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 10/04/2016] [Indexed: 11/26/2022] Open
Abstract
Background In malaria-endemic areas, the first exposure to malaria antigens often occurs in utero when the fetal immune system is poised towards the development of tolerance. Children exposed to placental malaria have an increased risk of clinical malaria in the first few years of life compared to unexposed children. Recent work has suggested the potential of pregnancy-associated malaria to induce immune tolerance in children living in malaria-endemic areas. A study was completed to evaluate the effect of malaria exposure during pregnancy on fetal immune tolerance and effector responses. Methods Using cord blood samples from a cohort of mother-infant pairs followed from early in pregnancy until delivery, flow cytometry analysis was completed to assess the relationship between pregnancy-associated malaria and fetal cord blood CD4 and dendritic cell phenotypes. Results Cord blood FoxP3+ Treg counts were higher in infants born to mothers with Plasmodium parasitaemia early in pregnancy (12–20 weeks of gestation; p = 0.048), but there was no association between Treg counts and the presence of parasites in the placenta at the time of delivery (by loop-mediated isothermal amplification (LAMP); p = 0.810). In contrast, higher frequencies of activated CD4 T cells (CD25+FoxP3−CD127+) were observed in the cord blood of neonates with active placental Plasmodium infection at the time of delivery (p = 0.035). This population exhibited evidence of effector memory differentiation, suggesting priming of effector T cells in utero. Lastly, myeloid dendritic cells were higher in the cord blood of infants with histopathologic evidence of placental malaria (p < 0.0001). Conclusion Together, these data indicate that in utero exposure to malaria drives expansion of both regulatory and effector T cells in the fetus, and that the timing of this exposure has a pivotal role in determining the polarization of the fetal immune response.
Collapse
|
21
|
Odorizzi PM, Feeney ME. Impact of In Utero Exposure to Malaria on Fetal T Cell Immunity. Trends Mol Med 2016; 22:877-888. [PMID: 27614925 DOI: 10.1016/j.molmed.2016.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 01/10/2023]
Abstract
Pregnancy-associated malaria, including placental malaria, causes significant morbidity and mortality worldwide. Recently, it has been suggested that in utero exposure of the fetus to malaria antigens may negatively impact the developing immune system and result in tolerance to malaria. Here, we review our current knowledge of fetal immunity to malaria, focusing on the dynamic interactions between maternal malaria infection, placental development, and the fetal immune system. A better understanding of the long-term impact of in utero malaria exposure on the development of natural immunity to malaria, immune responses to other childhood pathogens, and vaccine immunogenicity is urgently needed. This may guide the implementation of novel chemoprevention strategies during pregnancy and facilitate the push toward malaria vaccines.
Collapse
Affiliation(s)
- Pamela M Odorizzi
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, USA
| | - Margaret E Feeney
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
22
|
Zhang H. Progress in Research on the Mechanism and Treatment of Post-stroke Infection. INFECTION INTERNATIONAL 2016. [DOI: 10.1515/ii-2017-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractPost-stroke infection hinders the recovery of stroke patients and can even cause death. The main mechanism of post-stroke infection is related with the post-stroke center, the activation of the peripheral immune system, and the release of inflammatory factors caused by the lesion area and pathophysiological changes in the body. Therefore, elucidating the body’s abnormal immune inflammatory responses after stroke is crucial for the prevention, treatment, and alleviation of post-stroke infection.
Collapse
|
23
|
Sylvester B, Gasarasi DB, Aboud S, Tarimo D, Massawe S, Mpembeni R, Swedberg G. Prenatal exposure to Plasmodium falciparum increases frequency and shortens time from birth to first clinical malaria episodes during the first two years of life: prospective birth cohort study. Malar J 2016; 15:379. [PMID: 27448394 PMCID: PMC4957302 DOI: 10.1186/s12936-016-1417-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 07/04/2016] [Indexed: 11/23/2022] Open
Abstract
Background Prenatal exposure to Plasmodium falciparum affects development of protective immunity and susceptibility to subsequent natural challenges with similar parasite antigens. However, the nature of these effects has not been fully elucidated. The aim of this study was to determine the effect of prenatal exposure to P. falciparum on susceptibility to natural malaria infection, with a focus on median time from birth to first clinical malaria episode and frequency of clinical malaria episodes in the first 2 years of life. Methods A prospective birth cohort study was conducted in Rufiji district in Tanzania, between January 2013 and December 2015. Infants born to mothers with P. falciparum in the placenta at time of delivery were defined as exposed, and infants born to mothers without P. falciparum parasites in placenta were defined as unexposed. Placental infection was established by histological techniques. Out of 206 infants recruited, 41 were in utero exposed to P. falciparum and 165 infants were unexposed. All infants were monitored for onset of clinical malaria episodes in the first 2 years of life. The outcome measure was time from birth to first clinical malaria episode, defined by fever (≥37 °C) and microscopically determined parasitaemia. Median time to first clinical malaria episode between exposed and unexposed infants was assessed using Kaplan–Meier survival analysis and comparison was done by log rank. Association of clinical malaria episodes with prenatal exposure to P. falciparum was assessed by multivariate binary logistic regression. Comparative analysis of mean number of clinical malaria episodes between exposed and unexposed infants was done using independent sample t test. Results The effect of prenatal exposure to P. falciparum infection on clinical malaria episodes was statistically significant (Odds Ratio of 4.79, 95 % CI 2.21–10.38, p < 0.01) when compared to other confounding factors. Median time from birth to first clinical malaria episode for exposed and unexposed infants was 32 weeks (95 % CI 30.88–33.12) and 37 weeks (95 % CI 35.25–38.75), respectively, and the difference was statistically significant (p = 0.003). The mean number of clinical malaria episodes in exposed and unexposed infants was 0.51 and 0.30 episodes/infant, respectively, and the difference was statistically significant (p = 0.038). Conclusions Prenatal exposure to P. falciparum shortens time from birth to first clinical malaria episode and increases frequency of clinical malaria episodes in the first 2 years of life.
Collapse
Affiliation(s)
- Boniphace Sylvester
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania.
| | - Dinah B Gasarasi
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Donath Tarimo
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Siriel Massawe
- Department of Obstetrics and Gynaecology, School of Medicine, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Rose Mpembeni
- Department of Community Medicine, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Gote Swedberg
- Department of Medical Biochemistry, Biomedical Centre, Uppsala University, Uppsala, Sweden
| |
Collapse
|
24
|
Simon AK, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proc Biol Sci 2015; 282:20143085. [PMID: 26702035 PMCID: PMC4707740 DOI: 10.1098/rspb.2014.3085] [Citation(s) in RCA: 922] [Impact Index Per Article: 92.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/01/2015] [Indexed: 12/15/2022] Open
Abstract
This article reviews the development of the immune response through neonatal, infant and adult life, including pregnancy, ending with the decline in old age. A picture emerges of a child born with an immature, innate and adaptive immune system, which matures and acquires memory as he or she grows. It then goes into decline in old age. These changes are considered alongside the risks of different types of infection, autoimmune disease and malignancy.
Collapse
Affiliation(s)
- A Katharina Simon
- Nuffield Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Georg A Hollander
- Department of Paediatrics, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Andrew McMichael
- Nuffield Department of Medicine Research Building, University of Oxford, Old Road Campus, Oxford OX3 7FZ, UK
| |
Collapse
|
25
|
Nouatin O, Gbédandé K, Ibitokou S, Vianou B, Houngbegnon P, Ezinmegnon S, Borgella S, Akplogan C, Cottrell G, Varani S, Massougbodji A, Moutairou K, Troye-Blomberg M, Deloron P, Luty AJF, Fievet N. Infants' Peripheral Blood Lymphocyte Composition Reflects Both Maternal and Post-Natal Infection with Plasmodium falciparum. PLoS One 2015; 10:e0139606. [PMID: 26580401 PMCID: PMC4651557 DOI: 10.1371/journal.pone.0139606] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/14/2015] [Indexed: 11/19/2022] Open
Abstract
Maternal parasitoses modulate fetal immune development, manifesting as altered cellular immunological activity in cord blood that may be linked to enhanced susceptibility to infections in early life. Plasmodium falciparum typifies such infections, with distinct placental infection-related changes in cord blood exemplified by expanded populations of parasite antigen-specific regulatory T cells. Here we addressed whether such early-onset cellular immunological alterations persist through infancy. Specifically, in order to assess the potential impacts of P. falciparum infections either during pregnancy or during infancy, we quantified lymphocyte subsets in cord blood and in infants' peripheral blood during the first year of life. The principal age-related changes observed, independent of infection status, concerned decreases in the frequencies of CD4+, NKdim and NKT cells, whilst CD8+, Treg and Teff cells' frequencies increased from birth to 12 months of age. P. falciparum infections present at delivery, but not those earlier in gestation, were associated with increased frequencies of Treg and CD8+ T cells but fewer CD4+ and NKT cells during infancy, thus accentuating the observed age-related patterns. Overall, P. falciparum infections arising during infancy were associated with a reversal of the trends associated with maternal infection i.e. with more CD4+ cells, with fewer Treg and CD8+ cells. We conclude that maternal P. falciparum infection at delivery has significant and, in some cases, year-long effects on the composition of infants' peripheral blood lymphocyte populations. Those effects are superimposed on separate and independent age- as well as infant infection-related alterations that, respectively, either match or run counter to them.
Collapse
MESH Headings
- Adult
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Benin
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Female
- Fetal Blood/immunology
- Fetal Blood/parasitology
- Humans
- Immunophenotyping
- Infant
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lymphocyte Count
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/pathology
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/pathology
- Placenta/immunology
- Placenta/parasitology
- Placenta/pathology
- Plasmodium falciparum/immunology
- Pregnancy
- Pregnancy Complications, Parasitic/immunology
- Pregnancy Complications, Parasitic/parasitology
- Pregnancy Complications, Parasitic/pathology
- Retrospective Studies
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- Odilon Nouatin
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Komi Gbédandé
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Samad Ibitokou
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Bertin Vianou
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Parfait Houngbegnon
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Sem Ezinmegnon
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Sophie Borgella
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Institut de Recherche pour le Développement, MERIT UMR D216 Mère et enfant face aux infections tropicales, Paris, France
| | - Carine Akplogan
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Gilles Cottrell
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Institut de Recherche pour le Développement, MERIT UMR D216 Mère et enfant face aux infections tropicales, Paris, France
- PRES Sorbonne Paris Cité, Université Paris Descartes, Faculté de Pharmacie, Paris, France
| | - Stefania Varani
- Unit of Microbiology, Department of Diagnostic, Experimental and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Achille Massougbodji
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
| | - Kabirou Moutairou
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Marita Troye-Blomberg
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Philippe Deloron
- Institut de Recherche pour le Développement, MERIT UMR D216 Mère et enfant face aux infections tropicales, Paris, France
- PRES Sorbonne Paris Cité, Université Paris Descartes, Faculté de Pharmacie, Paris, France
| | - Adrian J. F. Luty
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Nadine Fievet
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Institut de Recherche pour le Développement, MERIT UMR D216 Mère et enfant face aux infections tropicales, Paris, France
- PRES Sorbonne Paris Cité, Université Paris Descartes, Faculté de Pharmacie, Paris, France
- * E-mail:
| |
Collapse
|
26
|
Lee HW, Kim TS, Kang YJ, Kim JY, Lee S, Lee WJ, Sohn Y, Lee HW. Up-regulated S100 calcium binding protein A8 in Plasmodium-infected patients correlates with CD4(+)CD25(+)Foxp3 regulatory T cell generation. Malar J 2015; 14:385. [PMID: 26438270 PMCID: PMC4594961 DOI: 10.1186/s12936-015-0855-4] [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: 06/30/2015] [Accepted: 08/22/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The pro-inflammatory S100 calcium binding protein A8 (S100A8) is elevated in the serum of patients with Plasmodium falciparum malaria, but its function in Plasmodium vivax malaria is not yet clear. This function was investigated in P. vivax-infected patients in this study. METHODS The level of S100A8 in the serum was measured with ELISA. Full amino acids of S100A8 were synthesized to verify the functions for maturation of immature dendritic cell (iDC) and evaluation of CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) generation by mature DC (mDC). RESULTS A higher amount of S100A8 was detected in vivax-infected patients (141.2 ± 61.849 ng/ml, n = 40) compared with normal control group (48.1 ± 27.384 ng/ml, n = 40). The level of S100A8 did not coincide with that of anti-malarial antibody measured by indirect fluorescent antibody test (IFAT) using parasite-infected red blood cells as antigen. Programmed death-ligand 1 (PD-L1) was up-regulated on the surface of iDCs following treatment with synthetic S100A8, not with synthetic MSP-1, AMA-1 and CSP, as compared to the expression seen for non-treated iDCs. The addition of red blood cells of infected patients to iDCs also elevated their surface expression of CD86. However, the serum levels of S100A8 decreased with increase in parasitaemia. DCs matured by sera containing S100A8 generated Treg cells from naïve T cells. The ratio of Treg cells generated was inversely proportional to the concentration of S100A8 in sera. CONCLUSIONS Treg cells suppress the activity of cytotoxic T cells, which kill malaria parasites; therefore, the up-regulation of S100A8 in malaria patients may contribute to pathogen immune escape or tolerance.
Collapse
Affiliation(s)
- Hyeong-Woo Lee
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| | - Tong-Soo Kim
- Department of Parasitology and Tropical Medicine, Inha Research Institute for Medical Sciences, Inha University School of Medicine, Incheon, 400-712, Republic of Korea.
| | - Yoon-Joong Kang
- Department of Biomedical Science, Jungwon University, Goesan, Chungbuk, 367-805, Republic of Korea.
| | - Jung-Yeon Kim
- Division of Malaria and Parasitic Disease, Korea National Institute of Health, Osong, 363-951, Republic of Korea.
| | - Sangeun Lee
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| | - Won-Ja Lee
- Division of Arbovirus, Korea National Institute of Health, Osong, 363-951, Republic of Korea.
| | - Youngjoo Sohn
- Department of Anatomy, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea.
| | - Hyeong-Woo Lee
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
27
|
Baker CAR, Swainson L, Lin DL, Wong S, Hartigan-O'Connor DJ, Lifson JD, Tarantal AF, McCune JM. Exposure to SIV in utero results in reduced viral loads and altered responsiveness to postnatal challenge. Sci Transl Med 2015; 7:300ra125. [PMID: 26268312 PMCID: PMC5100009 DOI: 10.1126/scitranslmed.aac5547] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
HIV disease progression appears to be driven by increased immune activation. Given observations that fetal exposure to infectious pathogens in utero can result in reduced immune responses, or tolerance, to those pathogens postnatally, we hypothesized that fetal exposure to HIV may render the fetus tolerant to the virus, thus reducing damage caused by immune activation during infection later in life. To test this hypothesis, fetal rhesus macaques (Macaca mulatta) were injected with the attenuated virus SIVmac1A11 in utero and challenged with pathogenic SIVmac239 1 year after birth. SIVmac1A11-injected animals had significantly reduced plasma RNA viral loads (P < 0.02) up to 35 weeks after infection. Generalized estimating equations analysis was performed to identify immunologic and clinical measurements associated with plasma RNA viral load. A positive association with plasma RNA viral load was observed with the proportion of CD8(+) T cells expressing the transcription factor, FoxP3, and the proportion of CD4(+) T cells producing the lymphoproliferative cytokine, IL-2. In contrast, an inverse relationship was found with the frequencies of circulating CD4(+) and CD8(+) T cells displaying intermediate expression of the proliferation marker, Ki-67. Animals exposed to simian immunodeficiency virus (SIV) in utero appeared to have enhanced SIV-specific immune responses, a lower proportion of CD8(+) T cells expressing the exhaustion marker PD-1, and more circulating TH17 cells than controls. Although the development of tolerance was not demonstrated, these data suggest that rhesus monkeys exposed to SIVmac1A11 in utero had distinct immune responses associated with the control of viral replication after postnatal challenge.
Collapse
Affiliation(s)
- Chris A R Baker
- Graduate Group in Infectious Diseases and Immunity, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA. Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Louise Swainson
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Din L Lin
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Samson Wong
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Dennis J Hartigan-O'Connor
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94110, USA. Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA. California National Primate Research Center, Davis, CA 95616, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702, USA
| | - Alice F Tarantal
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, California National Primate Research Center, Davis, CA 95616, USA. Department of Pediatrics, University of California, Davis, Davis, CA 95616, USA. Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA 95616, USA
| | - Joseph M McCune
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94110, USA.
| |
Collapse
|
28
|
Boyle MJ, Jagannathan P, Farrington LA, Eccles-James I, Wamala S, McIntyre TI, Vance HM, Bowen K, Nankya F, Auma A, Nalubega M, Sikyomu E, Naluwu K, Rek J, Katureebe A, Bigira V, Kapisi J, Tappero J, Muhindo MK, Greenhouse B, Arinaitwe E, Dorsey G, Kamya MR, Feeney ME. Decline of FoxP3+ Regulatory CD4 T Cells in Peripheral Blood of Children Heavily Exposed to Malaria. PLoS Pathog 2015; 11:e1005041. [PMID: 26182204 PMCID: PMC4504515 DOI: 10.1371/journal.ppat.1005041] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/23/2015] [Indexed: 12/27/2022] Open
Abstract
FoxP3+ regulatory CD4 T cells (Tregs) help to maintain the delicate balance between pathogen-specific immunity and immune-mediated pathology. Prior studies suggest that Tregs are induced by P. falciparum both in vivo and in vitro; however, the factors influencing Treg homeostasis during acute and chronic infections, and their role in malaria immunopathogenesis, remain unclear. We assessed the frequency and phenotype of Tregs in well-characterized cohorts of children residing in a region of high malaria endemicity in Uganda. We found that both the frequency and absolute numbers of FoxP3+ Tregs in peripheral blood declined markedly with increasing prior malaria incidence. Longitudinal measurements confirmed that this decline occurred only among highly malaria-exposed children. The decline of Tregs from peripheral blood was accompanied by reduced in vitro induction of Tregs by parasite antigen and decreased expression of TNFR2 on Tregs among children who had intense prior exposure to malaria. While Treg frequencies were not associated with protection from malaria, there was a trend toward reduced risk of symptomatic malaria once infected with P. falciparum among children with lower Treg frequencies. These data demonstrate that chronic malaria exposure results in altered Treg homeostasis, which may impact the development of antimalarial immunity in naturally exposed populations. In malaria endemic regions, immunity is slow to develop and does not provide substantial protection against reinfection. Rather, following repeated exposure, older children and adults eventually develop protection from most symptomatic manifestations of the infection. This may be due in part to the induction of immunoregulatory mechanisms by the P. falciparum parasite, such as FoxP3+ regulatory T cells (Tregs). Prior human studies have shown that Tregs are induced by malaria parasites both in vivo and in vitro, but the role of these cells in immunity in children who are chronically exposed to malaria remains unclear. In this study, we assessed the frequency and features of Tregs among children from areas of high malaria transmission in Uganda. We found that this regulatory T cell population declined markedly with increasing malaria episodes. This loss was associated with decreased expression of TNFR2, which is a protein implicated in stability of Tregs. Additionally, T cells from highly malaria exposed children demonstrated a reduced propensity to differentiate into Tregs following parasite stimulation. Together our data suggest that repeated episodes of malaria alter Treg homeostasis, which may influence the development of immunity to malaria in children.
Collapse
Affiliation(s)
- Michelle J. Boyle
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Center for Biomedical Research, The Burnet Institute, Melbourne, Australia
| | - Prasanna Jagannathan
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Lila A. Farrington
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Ijeoma Eccles-James
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Samuel Wamala
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Tara I McIntyre
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Hilary M. Vance
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Katherine Bowen
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | | | - Ann Auma
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Esther Sikyomu
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Kate Naluwu
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - John Rek
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Victor Bigira
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - James Kapisi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Mary K Muhindo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Bryan Greenhouse
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | | | - Grant Dorsey
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Moses R. Kamya
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Margaret E. Feeney
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
29
|
Yoon SH, Hur M, Hwang HS, Kwon HS, Sohn IS. The difference of lymphocyte subsets including regulatory T-cells in umbilical cord blood between AGA neonates and SGA neonates. Yonsei Med J 2015; 56:798-804. [PMID: 25837188 PMCID: PMC4397452 DOI: 10.3349/ymj.2015.56.3.798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE This study aimed to compare the regulatory T cells in cord blood of appropriate for gestational age (AGA) neonates with those of small for gestational age (SGA) neonates. MATERIALS AND METHODS Umbilical cord blood was collected upon labor in 108 healthy full-term (between 37 and 41 gestational weeks) neonates, who were born between November 2010 and April 2012. Among them, 77 samples were obtained from AGA neonates, and 31 samples were obtained from SGA neonates. Regulatory T cells and lymphocyte subsets were determined using a flow cytometer. Student's t-test for independent samples was used to compare differences between AGA and SGA neonates. RESULTS Regulatory T cells in cord blood were increased in the SGA group compared with normal controls (p=0.041). However, cytotoxic T cells in cord blood were significantly decreased in the SGA group compared with normal controls (p=0.007). CONCLUSION This is the first study to compare the distribution of lymphocyte subsets including regulatory T cells in cord blood between AGA neonates and SGA neonates.
Collapse
Affiliation(s)
- Sang Hee Yoon
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Mina Hur
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Han Sung Hwang
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Han Sung Kwon
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - In Sook Sohn
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea.
| |
Collapse
|
30
|
Malhotra I, McKibben M, Mungai P, McKibben E, Wang X, Sutherland LJ, Muchiri EM, King CH, King CL, LaBeaud AD. Effect of antenatal parasitic infections on anti-vaccine IgG levels in children: a prospective birth cohort study in Kenya. PLoS Negl Trop Dis 2015; 9:e0003466. [PMID: 25590337 PMCID: PMC4295886 DOI: 10.1371/journal.pntd.0003466] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 12/09/2014] [Indexed: 01/03/2023] Open
Abstract
Background Parasitic infections are prevalent among pregnant women in sub-Saharan Africa. We investigated whether prenatal exposure to malaria and/or helminths affects the pattern of infant immune responses to standard vaccinations against Haemophilus influenzae (Hib), diphtheria (DT), hepatitis B (Hep B) and tetanus toxoid (TT). Methods and Findings 450 Kenyan women were tested for malaria, schistosomiasis, lymphatic filariasis (LF), and intestinal helminths during pregnancy. After three standard vaccinations at 6, 10 and 14 weeks, their newborns were followed biannually to age 36 months and tested for absolute levels of IgG against Hib, DT, Hep B, and TT at each time point. Newborns’ cord blood (CB) lymphocyte responses to malaria blood-stage antigens, soluble Schistosoma haematobium worm antigen (SWAP), and filaria antigen (BMA) were also assessed. Three immunophenotype categories were compared: i) tolerant (those having Plasmodium-, Schistosoma-, or Wuchereria-infected mothers but lacking respective Th1/Th2-type recall responses at birth to malaria antigens, SWAP, or BMA); ii) sensitized (those with infected/uninfected mothers and detectable Th1/Th2-type CB recall response to respective parasite antigen); or iii) unexposed (no evidence of maternal infection or CB recall response). Overall, 78.9% of mothers were infected with LF (44.7%), schistosomiasis (32.4%), malaria (27.6%) or hookworm (33.8%). Antenatal maternal malaria, LF, and hookworm were independently associated with significantly lower Hib-specific IgG. Presence of multiple maternal infections was associated with lower infant IgG levels against Hib and DT antigens post-vaccination. Post-vaccination IgG levels were also significantly associated with immunophenotype: malaria-tolerized infants had reduced response to DT, whereas filaria-tolerized infants showed reduced response to Hib. Conclusions There is an impaired ability to develop IgG antibody responses to key protective antigens of Hib and diphtheria in infants of mothers infected with malaria and/or helminths during pregnancy. These findings highlight the importance of control and prevention of parasitic infections among pregnant women. Parasitic infections are prevalent among pregnant women in sub-Saharan Africa. Prenatal exposure to parasitic infections can generate several potential effects on fetal immune responses and affect functional antibody generation during subsequent vaccination. There is a paucity of data on the detrimental effects of chronic parasitic infections during pregnancy on the response to vaccine from birth to childhood. This paper highlights the overwhelming presence of helminth infection and malaria in pregnant women in rural Kenya. The study shows that the presence of single and multiple antenatal parasitic infections is associated with impaired infant IgG levels against Haemophilus influenzae (Hib) and diphtheria (DT) antigens post-vaccination from birth to 30 months of age. This study found that the response to DT was reduced in malaria-tolerized infants, and the response to Hib was impaired in filarial-tolerized infants; by contrast, the Schistosoma-tolerized group showed no effect. Deworming campaigns must be directed towards pregnant mothers, infants, and young children to improve response to vaccination.
Collapse
Affiliation(s)
- Indu Malhotra
- Case Western Reserve University, Center for Global Health and Diseases, Cleveland, Ohio, United States of America
- * E-mail:
| | - Maxim McKibben
- Case Western Reserve University, Center for Global Health and Diseases, Cleveland, Ohio, United States of America
| | - Peter Mungai
- Division of Vector Borne and Neglected Tropical Diseases, Ministry of Public Health and Sanitation, Nairobi, Kenya
| | - Elisabeth McKibben
- Case Western Reserve University, Center for Global Health and Diseases, Cleveland, Ohio, United States of America
| | - Xuelei Wang
- Case Western Reserve University, Clinical and Translational Science Collaborative, Cleveland, Ohio, United States of America
| | - Laura J. Sutherland
- Case Western Reserve University, Center for Global Health and Diseases, Cleveland, Ohio, United States of America
| | - Eric M. Muchiri
- Division of Vector Borne and Neglected Tropical Diseases, Ministry of Public Health and Sanitation, Nairobi, Kenya
| | - Charles H. King
- Case Western Reserve University, Center for Global Health and Diseases, Cleveland, Ohio, United States of America
| | - Christopher L. King
- Case Western Reserve University, Center for Global Health and Diseases, Cleveland, Ohio, United States of America
| | - A. Desiree LaBeaud
- Case Western Reserve University, Center for Global Health and Diseases, Cleveland, Ohio, United States of America
- Children’s Hospital Oakland Research Institute, Center for Immunobiology and Vaccine Development, Oakland, California, United States of America
| |
Collapse
|
31
|
Mother-to-Children Plasmodium falciparum Asymptomatic Malaria Transmission at Saint Camille Medical Centre in Ouagadougou, Burkina Faso. Malar Res Treat 2014; 2014:390513. [PMID: 25506464 PMCID: PMC4259075 DOI: 10.1155/2014/390513] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/01/2014] [Indexed: 12/04/2022] Open
Abstract
Background. Malaria's prevalence during pregnancy varies widely in parts of sub-Saharan Africa, including Burkina Faso. The objective of this study was to evaluate the incidence of mother-to-child malaria transmission during childbirth at St. Camille Medical Centre in the city of Ouagadougou. Methods. Two hundred and thirty-eight (238) women and their newborns were included in the study. Women consenting to participate in this study responded to a questionnaire that identified their demographic characteristics. Asymptomatic malaria infection was assessed by rapid detection test Acon (Acon Malaria Pf, San Diego, USA) and by microscopic examination of Giemsa-stained thick and thin smears from peripheral, placental, and umbilical cord blood. Birth weights were recorded and the biological analyses of mothers and newborns' blood were also performed. Results. The utilization of long-lasting insecticidal nets (LLINs) and intermittent preventive treatment with sulfadoxine-pyrimethamine (SP) were 86.6% and 84.4%, respectively. The parasitic infection rates of 9.5%, 8.9%, and 2.8% were recorded, respectively, for the peripheral, placental, and umbilical cord blood. Placental infection was strongly associated with the presence of parasites in the maternal peripheral blood and a parasite density of >1000 parasites/µL. Conclusion. The prevalence of congenital malaria was reduced but was associated with a high rate of mother-to-child malaria transmission.
Collapse
|
32
|
Randall LM, Hunter CA. Parasite dissemination and the pathogenesis of toxoplasmosis. Eur J Microbiol Immunol (Bp) 2014; 1:3-9. [PMID: 24466431 DOI: 10.1556/eujmi.1.2011.1.3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- L M Randall
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania Hill Pavilion, 380 South University Avenue, Philadelphia, PA 19146 USA
| | - C A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania Hill Pavilion, 380 South University Avenue, Philadelphia, PA 19146 USA
| |
Collapse
|
33
|
Cord blood CD4(+)CD25(+) regulatory T cells fail to inhibit cord blood NK cell functions due to insufficient production and expression of TGF-beta1. Cell Immunol 2014; 290:89-95. [PMID: 24950027 DOI: 10.1016/j.cellimm.2014.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/09/2014] [Accepted: 05/16/2014] [Indexed: 11/21/2022]
Abstract
Although CD4(+)CD25(+) Treg (Treg) cells are known to modulate NK cell functions, the modulation mechanism of these cells in cord blood has not been fully clarified. The purpose of this study was to clarify the mechanism whereby cord blood Treg cells modulate cord NK cells. By performing various cultures of purified NK cells with or without autologous Treg cells, diminished inhibitory effects of cord Treg cells towards cord NK cell functions, including activation, cytokine production, and cytotoxicity, were observed. We also observed lower secretion of sTGF-beta1 and lower expression of mTGF-beta1 by cord Treg cells than by adult Treg cells. These data revealed the capability of adult Treg cells to suppress rhIL-2-stimulated NK cell function by TGF-beta1, both membrane-bound and soluble types. The reduced inhibitory capabilities of cord Treg cells compared with adult Treg cells is thought to be due to insufficient expression of TGF-beta1.
Collapse
|
34
|
Scholzen A, Cooke BM, Plebanski M. Plasmodium falciparum induces Foxp3hi CD4 T cells independent of surface PfEMP1 expression via small soluble parasite components. Front Microbiol 2014; 5:200. [PMID: 24822053 PMCID: PMC4013457 DOI: 10.3389/fmicb.2014.00200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/15/2014] [Indexed: 01/08/2023] Open
Abstract
Elevated levels of regulatory T cells following Plasmodium infection are a well-reported phenomenon that can influence both protective and pathological anti-parasite responses, and might additionally impact on vaccine responses in acutely malaria infected individuals. The mechanisms underlying their induction or expansion by the parasite, however, are incompletely understood. In a previous study, Plasmodium falciparum infected red blood cells (iRBCs) were shown to induce effector-cytokine producing Foxp3int CD4+ T cells, as well as regulatory Foxp3hi CD4+ T cells in vitro. The aim of the present study was to determine the contribution of parasite components to the induction of Foxp3 expression in human CD4+ T cells. Using the surface PfEMP1-deficient parasite line 1G8, we demonstrate that induction of Foxp3hi and Foxp3int CD4+ T cells is independent of PfEMP1 expression on iRBCs. We further demonstrate that integrity of iRBCs is no requirement for the induction of Foxp3 expression. Finally, transwell experiments showed that induction of Foxp3 expression, and specifically the generation of Foxp3hi as opposed to Foxp3int CD4 T cells, can be mediated by soluble parasite components smaller than 20 nm and thus likely distinct from the malaria pigment hemozoin. These results suggest that the induction of Foxp3hi T cells by P. falciparum is largely independent of two key immune modulatory parasite components, and warrant future studies into the nature of the Foxp3hi inducing parasite components to potentially allow their exclusion from vaccine formulations.
Collapse
Affiliation(s)
- Anja Scholzen
- Department of Immunology, Monash University Melbourne, VIC, Australia ; Department of Medical Microbiology, Radboud University Medical Centre Nijmegen, Netherlands
| | - Brian M Cooke
- Department of Microbiology, Monash University Clayton, VIC, Australia
| | | |
Collapse
|
35
|
Abstract
Almost 7 million children under the age 5 die each year, and most of these deaths are attributable to vaccine-preventable infections. Young infants respond poorly to infections and vaccines. In particular, dendritic cells secrete less IL-12 and IL-18, CD8pos T cells and NK cells have defective cytolysis and cytokine production, and CD4pos T cell responses tend to bias towards a Th2 phenotype and promotion of regulatory T cells (Tregs). The basis for these differences is not well understood and may be in part explained by epigenetic differences, as well as immaturity of the infant's immune system. Here we present a third possibility, which involves active suppression by immune regulatory cells and place in context the immune suppressive pathways of mesenchymal stromal cells (MSC), myeloid-derived suppressor cells (MDSC), CD5pos B cells, and Tregs. The immune pathways that these immune regulatory cells inhibit are similar to those that are defective in the infant. Therefore, the immune deficiencies seen in infants could be explained, in part, by active suppressive cells, indicating potential new avenues for intervention.
Collapse
Affiliation(s)
- Ana L Gervassi
- Seattle Biomedical Research Institute and the University of Washington Departments of, Seattle WA
| | - Helen Horton
- Seattle Biomedical Research Institute and the University of Washington Departments of, Seattle WA ; Medicine, Seattle WA ; Global Health, Seattle WA
| |
Collapse
|
36
|
The management of paediatric allergy: not everybody's cup of tea--10-11th February 2012. Curr Opin Allergy Clin Immunol 2013; 13 Suppl 1:S1-50. [PMID: 23377496 DOI: 10.1097/aci.0b013e32835e8b94] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
37
|
Frosch AEP, John CC. Immunomodulation in Plasmodium falciparum malaria: experiments in nature and their conflicting implications for potential therapeutic agents. Expert Rev Anti Infect Ther 2013; 10:1343-56. [PMID: 23241191 DOI: 10.1586/eri.12.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Effective Plasmodium falciparum immunity requires a precisely timed and balanced response of inflammatory and anti-inflammatory immune regulators. These responses begin with innate immune effectors and are modulated over the course of an infection and between episodes to limit inflammation. To date, there are no effective immunomodulatory therapies for severe malaria. Some of the most potent immunomodulators are naturally occurring infections, including helminthic and chronic viral infections. This review examines malaria coinfection with these organisms, and their impact on malaria morbidity and immune responses. Overall, there is compelling evidence to suggest that chronic coinfections can modulate deleterious malaria-specific immune responses, suggesting that therapeutic agents may be effective if utilized early in infection. Examination of the mechanisms of these effects may serve as a platform to identify more targeted and effective malaria immunomodulatory therapeutics.
Collapse
Affiliation(s)
- Anne E P Frosch
- University of Minnesota, Center for Infectious Diseases and Microbiology Translational Research and Division of Global Pediatrics, McGuire Translational Research Facility, 2001 6th Street SE, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
38
|
Malaria modifies neonatal and early-life toll-like receptor cytokine responses. Infect Immun 2013; 81:2686-96. [PMID: 23690399 DOI: 10.1128/iai.00237-13] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Protection from infections in early life relies extensively on innate immunity, but it is unknown whether and how maternal infections modulate infants' innate immune responses, thereby altering susceptibility to infections. Plasmodium falciparum causes pregnancy-associated malaria (PAM), and epidemiological studies have shown that PAM enhances infants' susceptibility to infection with P. falciparum. We investigated how PAM-mediated exposures in utero affect innate immune responses and their relationship with infection in infancy. In a prospective study of mothers and their babies in Benin, we investigated changes in Toll-like receptor (TLR)-mediated cytokine responses related to P. falciparum infections. Whole-blood samples from 134 infants at birth and at 3, 6, and 12 months of age were stimulated with agonists specific for TLR3, TLR4, TLR7/8, and TLR9. TLR-mediated interleukin 6 (IL-6) and IL-10 production was robust at birth and then stabilized, whereas tumor necrosis factor alpha (TNF-α) and gamma interferon (IFN-γ) responses were weak at birth and then increased. In multivariate analyses, maternal P. falciparum infections at delivery were associated with significantly higher TLR3-mediated IL-6 and IL-10 responses in the first 3 months of life (P < 0.05) and with significantly higher TLR3-, TLR7/8-, and TLR9-mediated TNF-α responses between 6 and 12 months of age (P < 0.05). Prospective analyses showed that higher TLR3- and TLR7/8-mediated IL-10 responses at birth were associated with a significantly higher risk of P. falciparum infection in infancy (P < 0.05). Neonatal and infant intracellular TLR-mediated cytokine responses are conditioned by in utero exposure through PAM late in pregnancy. Enhanced TLR-mediated IL-10 responses at birth are associated with an increased risk of P. falciparum infection, suggesting a compromised ability to combat infection in early life.
Collapse
|
39
|
Abstract
The prevalence of food allergy has continued to rise over the last 10-15 years, with building concern over the underlying causes and the best strategies to reverse this. Although it is still not clear if infant feeding practices play any significant role in either the aetiology of this epidemic or in its prevention, these have nonetheless been core to many previous prevention strategies. Early 'allergen avoidance' strategies have not only failed, but have instead been increasingly associated with increased risk of allergic disease. Together with other observations in humans and animals, this suggests that earlier introduction of allergenic foods may be a more logical preventive strategy. Based on this, there are several randomised controlled trials world-wide assessing the merits of early introduction of complementary feeding and/or allergenic foods. Until the results of these studies are available it is difficult to provide definitive recommendations regarding the role of early feeding in the induction of oral tolerance and prevention of food allergy.
Collapse
|
40
|
Mueller I, Galinski MR, Tsuboi T, Arevalo-Herrera M, Collins WE, King CL. Natural acquisition of immunity to Plasmodium vivax: epidemiological observations and potential targets. ADVANCES IN PARASITOLOGY 2013; 81:77-131. [PMID: 23384622 DOI: 10.1016/b978-0-12-407826-0.00003-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Population studies show that individuals acquire immunity to Plasmodium vivax more quickly than Plasmodium falciparum irrespective of overall transmission intensity, resulting in the peak burden of P. vivax malaria in younger age groups. Similarly, actively induced P. vivax infections in malaria therapy patients resulted in faster and generally more strain-transcending acquisition of immunity than P. falciparum infections. The mechanisms behind the more rapid acquisition of immunity to P. vivax are poorly understood. Natural acquired immune responses to P. vivax target both pre-erythrocytic and blood-stage antigens and include humoral and cellular components. To date, only a few studies have investigated the association of these immune responses with protection, with most studies focussing on a few merozoite antigens (such as the Pv Duffy binding protein (PvDBP), the Pv reticulocyte binding proteins (PvRBPs), or the Pv merozoite surface proteins (PvMSP1, 3 & 9)) or the circumsporozoite protein (PvCSP). Naturally acquired transmission-blocking (TB) immunity (TBI) was also found in several populations. Although limited, these data support the premise that developing a multi-stage P. vivax vaccine may be feasible and is worth pursuing.
Collapse
Affiliation(s)
- Ivo Mueller
- Walter + Eliza Hall Institute, Infection & Immunity Division, Parkville, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Brambilla R, Couch Y, Lambertsen KL. The effect of stroke on immune function. Mol Cell Neurosci 2013; 53:26-33. [DOI: 10.1016/j.mcn.2012.08.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 06/27/2012] [Accepted: 08/22/2012] [Indexed: 02/09/2023] Open
|
42
|
Spence PJ, Langhorne J. T cell control of malaria pathogenesis. Curr Opin Immunol 2012; 24:444-8. [PMID: 22658628 DOI: 10.1016/j.coi.2012.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Accepted: 05/10/2012] [Indexed: 01/08/2023]
Abstract
Transmission of Plasmodium from mosquito to the mammalian host leads to a clinically silent pre-erythrocytic stage of malaria infection, and subsequent cyclical erythrocytic invasion associated with disease. Recent evidence demonstrates that it is the interplay between CD4+ and CD8+ T cells, and the regulation of their response, throughout infection that dictates immunity and the pathogenesis of malaria. The elicited T cell response is context dependent, influenced by diverse host and parasite factors, necessitating the development of a unifying model of T cell potential during Plasmodium infection. Only then can we predict their capacity to dictate the outcome of human disease.
Collapse
Affiliation(s)
- Philip J Spence
- Division of Parasitology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | | |
Collapse
|
43
|
Kim H, Moon HW, Hur M, Park CM, Yun YM, Hwang HS, Kwon HS, Sohn IS. Distribution of CD4+ CD25 high FoxP3+ regulatory T-cells in umbilical cord blood. J Matern Fetal Neonatal Med 2012; 25:2058-61. [PMID: 22348656 DOI: 10.3109/14767058.2012.666591] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE This study aimed to establish reference intervals for lymphocyte subsets including CD4(+)CD25(high)FoxP3(+) regulatory T-cells (Tregs) in umbilical cord blood. METHODS Umbilical cord blood was obtained after birth from 120 healthy full-term neonates, who were born between November 2010 and November 2011. Lymphocyte subsets including Tregs were analysed using flow cytometer (Beckman Coulter, Fullerton, CA, USA), and the reference intervals were defined using non-parametrical percentile methods according to the Clinical and Laboratory Standard Institute guideline (C28-A3). RESULTS The reference intervals for lymphocyte subsets were: helper T-cells (CD3(+)/CD4(+)), 15.40-70.06%; cytotoxic T-cells (CD3(+)/CD8(+)), 9.65-34.28%; B-cells (CD19(+)), 4.50-29.59%; and natural killer cells (CD3(-)/CD16(+)/CD56(+)), 1.42-28.03%. The reference interval for Tregs was 0.35-9.07%. CONCLUSIONS This study provides the reference intervals for lymphocyte subsets including Tregs in umbilical cord blood from healthy full-term neonates. These results could be used as fundamental data for clinical laboratory tests as well as future researches.
Collapse
Affiliation(s)
- Hanah Kim
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Glennie SJ, Nyirenda M, Williams NA, Heyderman RS. Do multiple concurrent infections in African children cause irreversible immunological damage? Immunology 2012; 135:125-32. [PMID: 22044389 DOI: 10.1111/j.1365-2567.2011.03523.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Much of the developing world, particularly sub-Saharan Africa, has high levels of morbidity and mortality associated with infectious diseases. The greatest risk of invasive disease is in the young, the malnourished and HIV-infected individuals. In many regions in Africa these vulnerable groups and the wider general population are under constant immune pressure from a range of environmental factors, under-nutrition and multiple concurrent infections from birth through to adulthood. Intermittent microbial exposure during childhood is required for the generation of naturally acquired immunity capable of protection against a range of infectious diseases in adult life. However, in the context of a resource-poor setting, the heavy burden of malarial, diarrhoeal and respiratory infections in childhood may subvert or suppress immune responses rather than protect, resulting in sub-optimal immunity. This review will explore how poor maternal health, HIV exposure, socio-economic and seasonal factors conspire to weaken childhood immune defences to disease and discuss the hypothesis that recurrent infections may drive immune dysregulation, leading to relative immune senescence and premature immunological aging.
Collapse
Affiliation(s)
- Sarah J Glennie
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi.
| | | | | | | |
Collapse
|
45
|
Lisciandro JG, Prescott SL, Nadal-Sims MG, Devitt CJ, Pomat W, Siba PM, Holt PG, Strickland D, van den Biggelaar AHJ. Comparison of neonatal T regulatory cell function in Papua New Guinean and Australian newborns. Pediatr Allergy Immunol 2012; 23:173-80. [PMID: 22192086 DOI: 10.1111/j.1399-3038.2011.01242.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Environmental changes, including declining microbial exposure, have been linked with the rising incidence of allergic and autoimmune diseases in 'western' populations. This potentially occurs by altering early development of immuno-regulatory pathways including T regulatory cells (T(reg)). There is now increasing evidence that such conditioning begins in utero. METHODS We compared neonatal T(reg) from children born under typical western conditions (Australia, AUS) with those of neonates born under more traditional conditions of high microbial burden (Papua New Guinea, PNG). RESULTS The frequency of neonatal T(reg), defined as CD4(+) Foxp3(+) CD127(-) CD25(+/high) was found to be higher in the cord blood of AUS compared to PNG newborns. However, cord T(reg) suppressive function in a small subset of children was qualitatively similar between PNG and AUS newborns in both a T(reg) depletion assay and a T(reg) supplementation assay. CONCLUSIONS These findings do not support the hypothesis that living in a 'western' versus more traditional environment leads to poor induction or suppressive function of neonatal T(reg). However, environmentally-induced immuno-regulation may potentially occur via alternative mechanisms in PNG newborns that should now be investigated further.
Collapse
Affiliation(s)
- Joanne G Lisciandro
- Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, Perth, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dauby N, Goetghebuer T, Kollmann TR, Levy J, Marchant A. Uninfected but not unaffected: chronic maternal infections during pregnancy, fetal immunity, and susceptibility to postnatal infections. THE LANCET. INFECTIOUS DISEASES 2012; 12:330-40. [PMID: 22364680 DOI: 10.1016/s1473-3099(11)70341-3] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic infections during pregnancy are highly prevalent in some parts of the world. Infections with helminths, Trypanosoma cruzi, Plasmodium spp, and HIV might affect the development of fetal immunity and susceptibility to postnatal infections independently of in-utero transmission of the pathogens. Fetal adaptive immune responses are common in neonates who have been exposed to maternal infection during pregnancy but not infected themselves. Such responses could affect the development of immunity to the homologous pathogens and their control during the first few years of life. Fetal innate and regulatory responses might also affect immunity to unrelated pathogens and responses to vaccines. Strategies to improve child health should integrate the possible clinical implications of in-utero exposure to chronic maternal infections.
Collapse
Affiliation(s)
- Nicolas Dauby
- Institute for Medical Immunology, Université Libre de Bruxelles, Charleroi, Belgium
| | | | | | | | | |
Collapse
|
47
|
Freitas do Rosário AP, Lamb T, Spence P, Stephens R, Lang A, Roers A, Muller W, O’Garra A, Langhorne J. IL-27 promotes IL-10 production by effector Th1 CD4+ T cells: a critical mechanism for protection from severe immunopathology during malaria infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 188:1178-90. [PMID: 22205023 PMCID: PMC3272378 DOI: 10.4049/jimmunol.1102755] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Infection with the malaria parasite, Plasmodium, is characterized by excessive inflammation. The establishment of a precise balance between the pro- and anti-inflammatory responses is critical to guarantee control of the parasite and survival of the host. IL-10, a key regulatory cytokine produced by many cells of the immune system, has been shown to protect mice against pathology during acute Plasmodium0 chabaudi chabaudi AS model of malaria. However, the critical cellular source of IL-10 is still unknown. In this article, we demonstrate that T cell-derived IL-10 is necessary for the control of pathology during acute malaria, as mice bearing specific deletion of Il10 in T cells fully reproduce the phenotype observed in Il10(-)(/)(-) mice, with significant weight loss, decline in temperature, and increased mortality. Furthermore, we show that IFN-γ(+) Th1 cells are the main producers of IL-10 throughout acute infection, expressing high levels of CD44 and ICOS, and low levels of CD127. Although Foxp3(+) regulatory CD4(+) T cells produce IL-10 during infection, highly activated IFN-γ(+) Th1 cells were shown to be the essential and sufficient source of IL-10 to guarantee protection against severe immune-mediated pathology. Finally, in this model of malaria, we demonstrate that the generation of protective IL10(+)IFN-γ(+) Th1 cells is dependent on IL-27 signaling and independent of IL-21.
Collapse
Affiliation(s)
| | - Tracey Lamb
- Divisions of Parasitology, MRC National Institute for Medical Research, London, UK
| | - Philip Spence
- Divisions of Parasitology, MRC National Institute for Medical Research, London, UK
| | - Robin Stephens
- Divisions of Parasitology, MRC National Institute for Medical Research, London, UK
| | - Agathe Lang
- Divisions of Parasitology, MRC National Institute for Medical Research, London, UK
| | - Axel Roers
- Medical Faculty Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Werner Muller
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Anne O’Garra
- Immunoregulation, MRC National Institute for Medical Research, London, UK
| | - Jean Langhorne
- Divisions of Parasitology, MRC National Institute for Medical Research, London, UK
| |
Collapse
|
48
|
Adalid-Peralta L, Fragoso G, Fleury A, Sciutto E. Mechanisms underlying the induction of regulatory T cells and its relevance in the adaptive immune response in parasitic infections. Int J Biol Sci 2011; 7:1412-26. [PMID: 22110392 PMCID: PMC3221948 DOI: 10.7150/ijbs.7.1412] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/01/2011] [Indexed: 12/22/2022] Open
Abstract
To fulfill its function, the immune system must detect and interpret a wide variety of signals and adjust the magnitude, duration, and specific traits of each response during the complex host-parasite relationships in parasitic infections. Inflammation must be tightly regulated since uncontrolled inflammation may be as destructive as the triggering stimulus and leads to immune-mediated tissue injury. During recent years, increasing evidence points to regulatory T cells (Tregs) as key anti-inflammatory cells, critically involved in limiting the inflammatory response. Herein, we review the published information on the induction of Tregs and summarize the most recent findings on Treg generation in parasitic diseases.
Collapse
|
49
|
Tweyongyere R, Mawa PA, Kihembo M, Jones FM, Webb EL, Cose S, Dunne DW, Vennervald BJ, Elliott AM. Effect of praziquantel treatment of Schistosoma mansoni during pregnancy on immune responses to schistosome antigens among the offspring: results of a randomised, placebo-controlled trial. BMC Infect Dis 2011; 11:234. [PMID: 21888656 PMCID: PMC3176493 DOI: 10.1186/1471-2334-11-234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 09/02/2011] [Indexed: 11/10/2022] Open
Abstract
Background Offspring of women with schistosomiasis may exhibit immune responsiveness to schistosomes due to in utero sensitisation or trans-placental transfer of antibodies. Praziquantel treatment during pregnancy boosts maternal immune responses to schistosome antigens and reduces worm burden. Effects of praziquantel treatment during pregnancy on responses among offspring are unknown. Methods In a trial of anthelminthic treatment during pregnancy in Uganda (ISRCTN32849447; http://www.controlled-trials.com/ISRCTN32849447/elliott), offspring of women with Schistosoma mansoni were examined for cytokine and antibody responses to schistosome worm (SWA) and egg (SEA) antigen, in cord blood and at age one year. Relationships to maternal responses and pre-treatment infection intensities were examined, and responses were compared between the offspring of women who did, or did not receive praziquantel treatment during pregnancy. Results Of 388 S. mansoni-infected women studied, samples were obtained at age one year from 215 of their infants. Stool examination for S. mansoni eggs was negative for all infants. Cord and infant samples were characterised by very low cytokine production in response to schistosome antigens with the exception of cord IL-10 responses, which were substantial. Cord and infant cytokine responses showed no association with maternal responses. As expected, cord blood levels of immunoglobulin (Ig) G to SWA and SEA were high and correlated with maternal antibodies. However, by age one year IgG levels had waned and were hardly detectable. Praziquantel treatment during pregnancy showed no effect on cytokine responses or antibodies levels to SWA or SEA either in cord blood or at age one year, except for IgG1 to SWA, which was elevated in infants of treated mothers, reflecting maternal levels. There was some evidence that maternal infection intensity was positively associated with cord blood IL-5 and IL-13 responses to SWA, and IL-5 responses to SEA, and that this association was modified by treatment with praziquantel. Conclusions Despite strong effects on maternal infection intensity and maternal immune responses, praziquantel treatment of infected women during pregnancy had no effect on anti-schistosome immune responses among offspring by age one year. Whether the treatment will impact upon the offspring's responses on exposure to primary schistosome infection remains to be elucidated. Trial registration ISRCTN: ISRCTN32849447
Collapse
|
50
|
Köhler C, Adegnika AA, van der Linden R, Luty AJF, Kremsner PG. Phenotypic characterization of mononuclear blood cells from pregnant Gabonese and their newborns. Trop Med Int Health 2011; 16:1061-9. [DOI: 10.1111/j.1365-3156.2011.02812.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|