1
|
Dzanibe S, Wilk AJ, Canny S, Ranganath T, Alinde B, Rubelt F, Huang H, Davis MM, Holmes SP, Jaspan HB, Blish CA, Gray CM. Premature skewing of T cell receptor clonality and delayed memory expansion in HIV-exposed infants. Nat Commun 2024; 15:4080. [PMID: 38744812 PMCID: PMC11093981 DOI: 10.1038/s41467-024-47955-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
While preventing vertical HIV transmission has been very successful, HIV-exposed uninfected infants (iHEU) experience an elevated risk to infections compared to HIV-unexposed and uninfected infants (iHUU). Here we present a longitudinal multimodal analysis of infant immune ontogeny that highlights the impact of HIV/ARV exposure. Using mass cytometry, we show alterations in T cell memory differentiation between iHEU and iHUU being significant from week 15 of life. The altered memory T cell differentiation in iHEU was preceded by lower TCR Vβ clonotypic diversity and linked to TCR clonal depletion within the naïve T cell compartment. Compared to iHUU, iHEU had elevated CD56loCD16loPerforin+CD38+CD45RA+FcεRIγ+ NK cells at 1 month postpartum and whose abundance pre-vaccination were predictive of vaccine-induced pertussis and rotavirus antibody responses post 3 months of life. Collectively, HIV/ARV exposure disrupted the trajectory of innate and adaptive immunity from birth which may underlie relative vulnerability to infections in iHEU.
Collapse
Affiliation(s)
- Sonwabile Dzanibe
- Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Aaron J Wilk
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Susan Canny
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA
- Division of Rheumatology, Department of Pediatrics, Seattle Children's Hospital, Seattle, WA, USA
| | - Thanmayi Ranganath
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Berenice Alinde
- Division of Immunology, Department of Biomedical Sciences, Biomedical Research Institute, Stellenbosch University, Cape Town, South Africa
| | - Florian Rubelt
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Huang Huang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA, USA
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Heather B Jaspan
- Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
- Seattle Children's Research Institute and Department of Paediatrics and Global Health, University of Washington, Seattle, WA, USA.
| | - Catherine A Blish
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA.
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Clive M Gray
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
- Division of Immunology, Department of Biomedical Sciences, Biomedical Research Institute, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
2
|
Camacho-Pacheco RT, Hernández-Pineda J, Brito-Pérez Y, Plazola-Camacho N, Coronado-Zarco IA, Arreola-Ramírez G, Bermejo-Haro MY, Najera-Hernández MA, González-Pérez G, Herrera-Salazar A, Olmos-Ortiz A, Soriano-Becerril D, Sandoval-Montes C, Figueroa-Damian R, Rodríguez-Martínez S, Mancilla-Herrera I. Disturbances in the IgG Antibody Profile in HIV-Exposed Uninfected Infants Associated with Maternal Factors. J Immunol Res 2024; 2024:8815767. [PMID: 38375063 PMCID: PMC10876311 DOI: 10.1155/2024/8815767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 02/21/2024] Open
Abstract
Over the last 20 years, the incidence of vertical HIV transmission has decreased from 25%-42% to less than 1%. Although there are no signs of infection, the health of HIV-exposed uninfected (HEU) infants is notoriously affected during the first months of life, with opportunistic infections being the most common disease. Some studies have reported effects on the vertical transfer of antibodies, but little is known about the subclass distribution of these antibodies. We proposed to evaluate the total IgG concentration and its subclasses in HIV+ mothers and HEU pairs and to determine which maternal factors condition their levels. In this study, plasma from 69 HEU newborns, their mothers, and 71 control pairs was quantified via immunoassays for each IgG isotype. Furthermore, we followed the antibody profile of HEUs throughout the first year of life. We showed that mothers present an antibody profile characterized by high concentrations of IgG1 and IgG3 but reduced IgG2, and HEU infants are born with an IgG subclass profile similar to that of their maternal pair. Interestingly, this passively transferred profile could remain influenced even during their own antibody production in HEU infants, depending on maternal conditions such as CD4+ T-cell counts and maternal antiretroviral treatment. Our findings indicate that HEU infants exhibit an altered IgG subclass profile influenced by maternal factors, potentially contributing to their increased susceptibility to infections.
Collapse
Affiliation(s)
- Rodrigo T. Camacho-Pacheco
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Jessica Hernández-Pineda
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Yesenia Brito-Pérez
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Noemi Plazola-Camacho
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | | | | | - Mextli Y. Bermejo-Haro
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - M. Angel Najera-Hernández
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Gabriela González-Pérez
- Department of Physiology and Cellular Development, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Alma Herrera-Salazar
- Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, UNAM, Cuautitlán Izcalli, Mexico
| | - Andrea Olmos-Ortiz
- Immunobiochemistry Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Diana Soriano-Becerril
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Claudia Sandoval-Montes
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ricardo Figueroa-Damian
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Sandra Rodríguez-Martínez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ismael Mancilla-Herrera
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| |
Collapse
|
3
|
Nyangahu DD, Happel AU, Wendoh J, Kiravu A, Wang Y, Feng C, Plumlee C, Cohen S, Brown BP, Djukovic D, Ganief T, Gasper M, Raftery D, Blackburn JM, Allbritton NL, Gray CM, Paik J, Urdahl KB, Jaspan HB. Bifidobacterium infantis associates with T cell immunity in human infants and is sufficient to enhance antigen-specific T cells in mice. SCIENCE ADVANCES 2023; 9:eade1370. [PMID: 38064556 PMCID: PMC10708209 DOI: 10.1126/sciadv.ade1370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/09/2023] [Indexed: 12/18/2023]
Abstract
Bacille Calmette-Guerin (BCG) vaccine can elicit good TH1 responses in neonates. We hypothesized that the pioneer gut microbiota affects vaccine T cell responses. Infants who are HIV exposed but uninfected (iHEU) display an altered immunity to vaccination. BCG-specific immune responses were analyzed at 7 weeks of age in iHEU, and responses were categorized as high or low. Bifidobacterium longum subsp. infantis was enriched in the stools of high responders, while Bacteroides thetaiotaomicron was enriched in low responders at time of BCG vaccination. Neonatal germ-free or SPF mice orally gavaged with live B. infantis exhibited significantly higher BCG-specific T cells compared with pups gavaged with B. thetaiotaomicron. B. infantis and B. thetaiotaomicron differentially affected stool metabolome and colonic transcriptome. Human colonic epithelial cells stimulated with B. infantis induced a unique gene expression profile versus B. thetaiotaomicron. We thus identified a causal role of B. infantis in early-life antigen-specific immunity.
Collapse
Affiliation(s)
- Donald D. Nyangahu
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Anna-Ursula Happel
- Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Jerome Wendoh
- Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Agano Kiravu
- Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Colin Feng
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Courtney Plumlee
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Sara Cohen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Bryan P. Brown
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Danijel Djukovic
- Northwest Metabolomics Research Center, University of Washington, Seattle, WA, USA
| | - Tariq Ganief
- Institute of Infectious Diseases and Molecular Medicine, Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, University of Cape Town, Cape Town, South Africa
| | - Melanie Gasper
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, University of Washington, Seattle, WA, USA
| | - Jonathan M. Blackburn
- Institute of Infectious Diseases and Molecular Medicine, Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, University of Cape Town, Cape Town, South Africa
| | | | - Clive M. Gray
- Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Division of Immunology, University of Cape Town, Cape Town, South Africa
- Biomedical Research Institute, Division of Molecular Biology and Human Genetics, Stellenbosch University, Cape Town, South Africa
| | - Jisun Paik
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Kevin B. Urdahl
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, School of Medicine, University of Washington, Seattle WA, USA
| | - Heather B. Jaspan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Division of Immunology, University of Cape Town, Cape Town, South Africa
- Department of Pediatrics, School of Medicine, University of Washington, Seattle WA, USA
| |
Collapse
|
4
|
Wang X, Zhao Y, Bai T, Ye J, Lu S, Wu F, Li L, Chen J. Serum immune biomarker levels combined with hepatitis B virus infection status predict early recurrence of early-stage hepatocellular carcinoma with microvascular invasion after liver resection. Acta Chir Belg 2023; 123:659-665. [PMID: 36222747 DOI: 10.1080/00015458.2022.2136051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/09/2022] [Indexed: 11/13/2022]
Abstract
INTRODUCTION The tumor immune response plays a vital role in cancer recurrence in patients with malignancies. We aim to clarify the risk factors for early recurrence and investigate the efficacy of blood-based biomarkers to predict the risk of early recurrence in early-stage hepatocellular carcinoma (HCC) patients with microvascular invasion (MVI) after hepatectomy. MATERIALS AND METHODS A total of 101 cases of HCC with MVI who underwent liver resection were enrolled. Univariate and multivariate logistic regression analyses were performed to identify independent risk factors of early recurrence. We calculated the area under the receiver operating characteristic curve to evaluate the performance of the four biomarkers identified as risk factors for early recurrence. RESULTS Multiple logistic regression analysis indicated that complement (C)4, cluster of differentiation (CD)4+, immunoglobulin A (IgA), and hepatitis B virus (HBV) DNA of greater than 500 IU/mL were correlated with early recurrence of HCC. The area under the curve was greater for the combination model than for the HBV DNA, CD4+, IgA, or C4 models alone. CONCLUSION Preoperative serum CD4+, C4, IgA, and HBV DNA levels were linked with early recurrence of early-stage HCC with MVI and the combination model was of considerable predictive value for the prognosis of HCC with MVI.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Hepatobiliary Surgery, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China
| | - Yuanquan Zhao
- Department of Hepatobiliary Surgery, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China
| | - Tao Bai
- Department of Hepatobiliary Surgery, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China
| | - Jiazhou Ye
- Department of Hepatobiliary Surgery, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China
| | - Shaolong Lu
- Department of Hepatobiliary Surgery, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China
| | - Feixiang Wu
- Department of Hepatobiliary Surgery, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China
| | - Lequn Li
- Department of Hepatobiliary Surgery, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China
| | - Jie Chen
- Department of Hepatobiliary Surgery, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China
| |
Collapse
|
5
|
Floridia M, Orlando S, Andreotti M, Mphwere R, Kavalo T, Ciccacci F, Scarcella P, Marazzi MC, Giuliano M. A 12-month Prospective Study of HIV-infected and HIV-uninfected Women and Their Infants in Malawi: Comparative Analysis of Clinical Events and Infant Growth. Am J Trop Med Hyg 2023; 108:394-402. [PMID: 36535249 PMCID: PMC9896343 DOI: 10.4269/ajtmh.22-0482] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/16/2022] [Indexed: 12/23/2022] Open
Abstract
Few studies have compared clinical outcomes in HIV-exposed uninfected (HEU) and HIV-unexposed uninfected (HUU) infants in the current scenario of universal and lifelong maternal antiretroviral therapy (ART). HIV-uninfected and HIV-infected Malawian women receiving ART and their breastfed infants were followed for 12 months postpartum, analyzing the rates of infectious and noninfectious events and assessing infant growth at 6 weeks, 6 months, and 12 months. The cohorts included 227 mothers (70 HIV-negative, 157 HIV-positive) and 235 infants (72 HUU, 163 HEU). No maternal or infant deaths occurred during follow-up. HIV-negative women were less likely to complete follow-up (48.6% versus 91.1%). Mothers with and without HIV had similar rates of both infectious and noninfectious events per person-month. Infants who were HEU, compared with HUU, had higher rates of events of any type, lower respiratory tract infections (LRTI), and noninfectious events. HEU had lower body mass index (BMI) at 6 weeks but did not differ from HUU in all anthropometric measures at 6 and 12 months; in growth between 6 weeks and 12 months; and in occurrence of stunting, underweight, and wasting at 6 weeks, 6 months, and 12 months. During the first year of life, infants who were HEU, compared with HUU, showed a transiently lower BMI and an increased risk of LRTI.
Collapse
Affiliation(s)
- Marco Floridia
- Istituto Superiore di Sanità, National Center for Global Health, Rome, Italy
| | - Stefano Orlando
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Mauro Andreotti
- Istituto Superiore di Sanità, National Center for Global Health, Rome, Italy
| | | | - Thom Kavalo
- DREAM Program, Community of S. Egidio, Blantyre, Malawi
| | - Fausto Ciccacci
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Paola Scarcella
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Marina Giuliano
- Istituto Superiore di Sanità, National Center for Global Health, Rome, Italy
| |
Collapse
|
6
|
du Toit LDV, Prinsloo A, Steel HC, Feucht U, Louw R, Rossouw TM. Immune and Metabolic Alterations in Children with Perinatal HIV Exposure. Viruses 2023; 15:v15020279. [PMID: 36851493 PMCID: PMC9966389 DOI: 10.3390/v15020279] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
With the global rollout of mother-to-child prevention programs for women living with HIV, vertical transmission has been all but eliminated in many countries. However, the number of children who are exposed in utero to HIV and antiretroviral therapy (ART) is ever-increasing. These children who are HIV-exposed-but-uninfected (CHEU) are now well recognized as having persistent health disparities compared to children who are HIV-unexposed-and-uninfected (CHUU). Differences reported between these two groups include immune dysfunction and higher levels of inflammation, cognitive and metabolic abnormalities, as well as increased morbidity and mortality in CHEU. The reasons for these disparities remain largely unknown. The present review focuses on a proposed link between immunometabolic aberrations and clinical pathologies observed in the rapidly expanding CHEU population. By drawing attention, firstly, to the significance of the immune and metabolic alterations observed in these children, and secondly, the impact of their healthcare requirements, particularly in low- and middle-income countries, this review aims to sensitize healthcare workers and policymakers about the long-term risks of in utero exposure to HIV and ART.
Collapse
Affiliation(s)
- Louise D V du Toit
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
- UP Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria 0001, South Africa
- Maternal and Infant Health Care Strategies Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
| | - Andrea Prinsloo
- UP Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria 0001, South Africa
- Maternal and Infant Health Care Strategies Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Hematology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Helen C Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Ute Feucht
- UP Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria 0001, South Africa
- Maternal and Infant Health Care Strategies Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Pediatrics, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Theresa M Rossouw
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
- UP Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria 0001, South Africa
- Maternal and Infant Health Care Strategies Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
| |
Collapse
|
7
|
Gee S, Chandiramani M, Seow J, Pollock E, Modestini C, Das A, Tree T, Doores KJ, Tribe RM, Gibbons DL. The legacy of maternal SARS-CoV-2 infection on the immunology of the neonate. Nat Immunol 2021; 22:1490-1502. [PMID: 34616036 DOI: 10.1038/s41590-021-01049-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Despite extensive studies into severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the effect of maternal infection on the neonate is unclear. To investigate this, we characterized the immunology of neonates born to mothers with confirmed SARS-CoV-2 infection during pregnancy. Here we show that maternal SARS-CoV-2 infection affects the neonatal immune system. Despite similar proportions of B cells, CD4+ T cells and CD8+ T cells, increased percentages of natural killer cells, Vδ2+ γδ T cells and regulatory T cells were detected in neonates born to mothers with recent or ongoing infection compared with those born to recovered or uninfected mothers. Increased plasma cytokine levels were also evident in neonates and mothers within the recent or ongoing infection group. Cytokine functionality was enhanced in neonates born to SARS-CoV-2-exposed mothers, compared to those born to uninfected mothers. In most neonates, this immune imprinting was nonspecific, suggesting vertical transmission of SARS-CoV-2 is limited, a finding supported by a lack of SARS-CoV-2-specific IgM in neonates despite maternal IgG transfer.
Collapse
Affiliation(s)
- Sarah Gee
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Manju Chandiramani
- Department of Women's Health, Guy's and St Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Jeffrey Seow
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Emily Pollock
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Carlotta Modestini
- Department of Women's Health, Guy's and St Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Abhishek Das
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Timothy Tree
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Katie J Doores
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Rachel M Tribe
- Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, St Thomas' Hospital, London, UK
| | - Deena L Gibbons
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK.
| |
Collapse
|
8
|
Brito-Pérez Y, Camacho-Pacheco RT, Plazola-Camacho N, Soriano-Becerril D, Coronado-Zarco IA, Arreola-Ramírez G, González-Pérez G, Herrera-Salazar A, Flores-González J, Bermejo-Haro MY, Casorla-Cervantes BG, Soto-López IA, Hernández-Pineda J, Sandoval-Montes C, Rodríguez-Martínez S, Figueroa-Damian R, Mancilla-Herrera I. Impaired T helper cell responses in human immunodeficiency virus-exposed uninfected newborns. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1541-1553. [PMID: 34409752 PMCID: PMC8589403 DOI: 10.1002/iid3.507] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022]
Abstract
Introduction HIV‐exposed uninfected (HEU) newborns suffer from higher risks of opportunistic infections during the first months of life compared to HIV‐unexposed uninfected (HUU) newborns. Alterations in thymic mass, amounts of T helper (Th) cells, T‐cell receptor diversity, and activation markers have been found in HEU newborns, suggesting alterations in T cell ontogeny and differentiation. However, little is known about the ability of these cells to produce specialized Th responses from CD4+ T cells. Method To characterize the Th cell profile, we evaluated the frequency of Th1 (CD183+CD194−CD196−/CXCR3+CCR4−CCR6−), Th2 (CD183−CD194+CD196−/CXCR3−CCR4+CCR6−), Th17 (CD183−CD194+CD196+/CXCR3−CCR4+CCR6+), and CD4+CD25++ blood T‐cell phenotypes in 50 HEU and 25 HUU newborns. Early activation markers on CD4+ T cells and the Th cytokine profile produced from mononuclear cells under polyclonal T cell stimulation were also studied. Additionally, we probed the ability of CD4+ T cells to differentiate into interferon (IFN)‐γ‐producing Th1 CD4+ T cells in vitro. Results Lower percentages of differentiated Th1, Th2, Th17, and CD4+CD25++ T cells were found in blood from HEU newborns than in blood from HUU newborns. However, polyclonally stimulated Th cells showed a similar ability to express CD69 and CD279 but produced less secreted interleukin (IL)‐2 and IL‐4. Interestingly, under Th1 differentiation conditions, the percentages of CD4+IFN‐γ+ T cells and soluble IFN‐γ were higher in HEU newborns than in HUU newborns. Conclusion HEU neonates are born with reduced proportions of differentiated Th1/Th2/Th17 and CD4+CD25++ T cells, but the intrinsic abilities of CD4+ T cells to acquire a Th1 profile are not affected by the adverse maternal milieu during development.
Collapse
Affiliation(s)
- Yesenia Brito-Pérez
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico.,Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México.,Posgrado en Inmunología, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rodrigo T Camacho-Pacheco
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico.,Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México.,Posgrado en Inmunología, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Noemi Plazola-Camacho
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Diana Soriano-Becerril
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Irma A Coronado-Zarco
- Neonatology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | | | - Gabriela González-Pérez
- Department of Physiology and Cellular Development, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Alma Herrera-Salazar
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Julio Flores-González
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Mextli Y Bermejo-Haro
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico.,Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México.,Posgrado en Inmunología, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Ismael A Soto-López
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Jessica Hernández-Pineda
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Claudia Sandoval-Montes
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Sandra Rodríguez-Martínez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Ricardo Figueroa-Damian
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Ismael Mancilla-Herrera
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| |
Collapse
|
9
|
Abstract
The neonatal period and early infancy are times of increased vulnerability to infection. The immune system of infants undergoes rapid changes and a number of factors can influence the maturation and function of the early infant immune system, amongst these factors are maternal infections and immunity. Infants who are HIV-exposed, but uninfected show important immune alterations, which are likely to be associated with the increased morbidity and mortality observed in these infants. Maternally derived antibodies are crucial in early life to protect infants from infection during the time when their own immune system is becoming more experienced and fully mature. However, maternal antibodies can also interfere with the infant's own antibody responses to primary vaccination. Preterm infants are particularly vulnerable to infection, having not had the opportunity to benefit from the transplacental transfer of maternal antibodies in late pregnancy. In addition, further differences have been observed in the innate and adaptive immune system between preterm and term infants. Here, we focus on maternal influences on the infant immune system, using HIV and maternal vaccination as examples and finish by considering how prematurity impacts infant immune responses to vaccination.
Collapse
Affiliation(s)
- Petra Zimmermann
- From the Department of Paediatrics, Fribourg Hospital HFR and Faculty of Science and Medicine, University of Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia
| | - Christine E Jones
- Faculty of Medicine and Institute for Life Sciences, University of Southampton and NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| |
Collapse
|
10
|
Wood MP, Wood LF, Templeton M, Fisher B, Lippy A, Jones CI, Lindestam Arlehamn CS, Sette A, Fuller JT, Murapa P, Jaspan HB, Fuller DH, Sodora DL. Transient Immune Activation in BCG-Vaccinated Infant Rhesus Macaques Is Not Sufficient to Influence Oral Simian Immunodeficiency Virus Infection. J Infect Dis 2021; 222:44-53. [PMID: 31605528 DOI: 10.1093/infdis/jiz382] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022] Open
Abstract
BCG vaccination has been demonstrated to increase levels of activated CD4+ T cells, thus potentially influencing mother-to-child transmission of human immunodeficiency virus (HIV). To assess the risk of BCG vaccination in HIV infection, we randomly assigned newborn rhesus macaques to receive BCG vaccine or remain unvaccinated and then undergo oral simian immunodeficiency virus (SIV) challenges 3 weeks later. We observed elevated levels of activated peripheral CD4+ T cells (ie, HLA-DR+CD38+CCR5+ CD4+ T cells) by week 3 after vaccination. BCG was also associated with an altered immune gene expression profile, as well as with monocyte activation in both peripheral blood and the draining axillary lymph node, indicating significant BCG vaccine-induced immune activation. Despite these effects, BCG vaccination did not increase the rate of SIV oral transmission or disease progression. Our findings therefore identify patterns of T-cell and monocyte activation that occur after BCG vaccination but do not support the hypothesis that BCG vaccination is a risk factor for postnatal HIV transmission or increased pathogenesis in infants.
Collapse
Affiliation(s)
- Matthew P Wood
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| | - Lianna F Wood
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| | - Megan Templeton
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| | - Bridget Fisher
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| | - Adriana Lippy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| | - Chloe I Jones
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, San Diego.,Department of Medicine, University of California-San Diego, La Jolla, California
| | - James T Fuller
- Department of Microbiology, Seattle, Washington; and University of Washington.,Washington National Primate Research Center, Seattle, Washington
| | - Patience Murapa
- Department of Microbiology, Seattle, Washington; and University of Washington.,Washington National Primate Research Center, Seattle, Washington
| | - Heather B Jaspan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| | - Deborah H Fuller
- Department of Microbiology, Seattle, Washington; and University of Washington.,Washington National Primate Research Center, Seattle, Washington
| | - Donald L Sodora
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
11
|
Smith C, Moraka NO, Ibrahim M, Moyo S, Mayondi G, Kammerer B, Leidner J, Gaseitsiwe S, Li S, Shapiro R, Lockman S, Weinberg A. Human Immunodeficiency Virus Exposure but Not Early Cytomegalovirus Infection Is Associated With Increased Hospitalization and Decreased Memory T-Cell Responses to Tetanus Vaccine. J Infect Dis 2020; 221:1167-1175. [PMID: 31711179 PMCID: PMC7075416 DOI: 10.1093/infdis/jiz590] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/07/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-exposed, uninfected (HEU) infants experience high rates of infectious morbidity. We hypothesized that early cytomegalovirus (CMV) infection was associated with increased hospitalization rates and decreased vaccine responses in HEU compared with HIV-unexposed (HUU) infants. METHODS Among infants enrolled in the Tshipidi study in Botswana, we determined CMV infection status by 6 months of age and compared hospitalization rates and responses to tetanus and Bacille Calmette-Guérin vaccines among HEU and HUU vaccinees. RESULTS Fifteen of 226 (6.6%) HEU infants and 17 (19.3%) of 88 HUU infants were CMV-infected by 6 months. The HEU infants were approximately 3 times as likely to be hospitalized compared with HUU infants (P = .02). The HEU peripheral blood cells produced less interleukin (IL)-2 (P = .004), but similar amounts of interferon-γ, after stimulation with tetanus toxoid. Antitetanus immunoglobulin G titers were similar between groups. Cellular responses to purified protein derivative stimulation did not differ between groups. Maternal receipt of 3-drug antiretroviral therapy compared with zidovudine was associated with increased IL-2 expression after tetanus toxoid stimulation. The infants' CMV infection status was not associated with clinical or vaccine response outcomes. CONCLUSIONS We observed that increased rates of hospitalization and decreased memory T-cell responses to tetanus vaccine were associated with HIV exposure and incomplete treatment of maternal HIV infection, but not early CMV infection.
Collapse
Affiliation(s)
- Christiana Smith
- University of Colorado School of Medicine, Denver, Colorado, USA
| | - Natasha O Moraka
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Stellenbosch University, Stellenbosch, South Africa
| | | | - Sikhulile Moyo
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Gloria Mayondi
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Betsy Kammerer
- Boston Children’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Jean Leidner
- Goodtables Data Consulting, Norman, Oklahoma, USA
| | - Simani Gaseitsiwe
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Shaobing Li
- University of Colorado School of Medicine, Denver, Colorado, USA
| | - Roger Shapiro
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Shahin Lockman
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Adriana Weinberg
- University of Colorado School of Medicine, Denver, Colorado, USA
| |
Collapse
|
12
|
Birungi N, Fadnes LT, Engebretsen IMS, Lie SA, Tumwine JK, Åstrøm AN. Association of maternal HIV-1 severity with dental caries: an observational study of uninfected 5- to 7-yr-old children of HIV-1-infected mothers without severe immune suppression. Eur J Oral Sci 2020; 128:46-54. [PMID: 31994250 DOI: 10.1111/eos.12669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2019] [Indexed: 12/13/2022]
Abstract
Treatment programs to prevent perinatal and postnatal HIV-1 transmission have become available in sub-Saharan Africa, leading to an emerging population of HIV-1 exposed uninfected (HEU) children. Exposure to HIV-1 in utero and during breastfeeding may increase the risk of morbidity and mortality in HEU children. This study estimated the association of the severity of maternal HIV-1 infection as assessed by CD4 count and viral load at baseline (7 d postpartum), with dmft count of their 5- to 7-yr-old HEU offspring. A follow-up study was conducted of HIV-1-infected mother-HEU children pairs (n = 164) from the Ugandan site of the ANRS 12341-PROMISE- PEP trial (ClinicalTrials.gov, number NCT00640263). HIV-1-infected mothers were interviewed and the HEU children were examined for caries using the World Health Organization's survey methods for field conditions and the dmft index. Directed acyclic graphs and negative binomial regression were used for analyses. The prevalence of 1 or more dmft was 48%. Negative binomial regression showed no association between the dmft count and maternal CD4 counts 7 d postpartum but a 10% lower dmft count with longer breastfeeding duration was found. Maternal CD4 count at birth was not associated with the dental caries experience in uninfected children born to women without severe immune suppression, while there appeared to be a protective effect of high viral load and breastfeeding duration.
Collapse
Affiliation(s)
- Nancy Birungi
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Lars Thore Fadnes
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway.,Bergen Addiction Research Group, Department of Addiction Medicine, Haukeland University Hospital, Bergen, Norway
| | | | - Stein Atle Lie
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - James Kashyugyera Tumwine
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University Kampala, Kampala, Uganda
| | | | | |
Collapse
|
13
|
Musimbi ZD, Rono MK, Otieno JR, Kibinge N, Ochola-Oyier LI, de Villiers EP, Nduati EW. Peripheral blood mononuclear cell transcriptomes reveal an over-representation of down-regulated genes associated with immunity in HIV-exposed uninfected infants. Sci Rep 2019; 9:18124. [PMID: 31792230 PMCID: PMC6889308 DOI: 10.1038/s41598-019-54083-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022] Open
Abstract
HIV-exposed uninfected (HEU) infants are disproportionately at a higher risk of morbidity and mortality, as compared to HIV-unexposed uninfected (HUU) infants. Here, we used transcriptional profiling of peripheral blood mononuclear cells to determine immunological signatures of in utero HIV exposure. We identified 262 differentially expressed genes (DEGs) in HEU compared to HUU infants. Weighted gene co-expression network analysis (WGCNA) identified six modules that had significant associations with clinical traits. Functional enrichment analysis on both DEGs and the six significantly associated modules revealed an enrichment of G-protein coupled receptors and the immune system, specifically affecting neutrophil function and antibacterial responses. Additionally, malaria pathogenicity genes (thrombospondin 1-(THBS 1), interleukin 6 (IL6), and arginine decarboxylase 2 (ADC2)) were down-regulated. Of interest, the down-regulated immunity genes were positively correlated to the expression of epigenetic factors of the histone family and high-mobility group protein B2 (HMGB2), suggesting their role in the dysregulation of the HEU transcriptional landscape. Overall, we show that genes primarily associated with neutrophil mediated immunity were repressed in the HEU infants. Our results suggest that this could be a contributing factor to the increased susceptibility to bacterial infections associated with higher morbidity and mortality commonly reported in HEU infants.
Collapse
Affiliation(s)
- Zaneta D Musimbi
- Center of Biotechnology and Bioinformatics, Chiromo Campus, University of Nairobi, Nairobi, Kenya.
| | - Martin K Rono
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya.
| | | | | | - Lynette Isabella Ochola-Oyier
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya
| | - Etienne Pierre de Villiers
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Eunice W Nduati
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya
| |
Collapse
|
14
|
Kiravu A, Osawe S, Happel AU, Nundalall T, Wendoh J, Beer S, Dontsa N, Alinde OB, Mohammed S, Datong P, Cameron DW, Rosenthal K, Abimiku A, Jaspan HB, Gray CM. Bacille Calmette-Guérin Vaccine Strain Modulates the Ontogeny of Both Mycobacterial-Specific and Heterologous T Cell Immunity to Vaccination in Infants. Front Immunol 2019; 10:2307. [PMID: 31649662 PMCID: PMC6793433 DOI: 10.3389/fimmu.2019.02307] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/12/2019] [Indexed: 01/24/2023] Open
Abstract
Differences in Bacille Calmette-Guérin (BCG) immunogenicity and efficacy have been reported, but various strains of BCG are administered worldwide. Since BCG immunization may also provide protection against off-target antigens, we sought to identify the impact of different BCG strains on the ontogeny of vaccine-specific and heterologous vaccine immunogenicity in the first 9 months of life, utilizing two African birth cohorts. A total of 270 infants were studied: 84 from Jos, Nigeria (vaccinated with BCG-Bulgaria) and 187 from Cape Town, South Africa (154 vaccinated with BCG-Denmark and 33 with BCG-Russia). Infant whole blood was taken at birth, 7, 15, and 36 weeks and short-term stimulated (12 h) in vitro with BCG, Tetanus and Pertussis antigens. Using multiparameter flow cytometry, CD4+ T cell memory subset polyfunctionality was measured by analyzing permutations of TNF-α, IL-2, and IFN-γ expression at each time point. Data was analyzed using FlowJo, SPICE, R, and COMPASS. We found that infants vaccinated with BCG-Denmark mounted significantly higher frequencies of BCG-stimulated CD4+ T cell responses, peaking at week 7 after immunization, and possessed durable polyfunctional CD4+ T cells that were in a more early differentiated memory stage when compared with either BCG-Bulgaria and BCG-Russia strains. The latter responses had lower polyfunctional scores and tended to accumulate in a CD4+ T cell naïve-like state (CD45RA+CD27+). Notably, BCG-Denmark immunization resulted in higher magnitudes and polyfunctional cytokine responses to heterologous vaccine antigens (Tetanus and Pertussis). Collectively, our data show that BCG strain was the strongest determinant of both BCG-stimulated and heterologous vaccine stimulated T cell magnitude and polyfunctionality. These findings have implications for vaccine policy makers, manufacturers and programs worldwide and also suggest that BCG-Denmark, the first vaccine received in many African infants, has both specific and off-target effects in the first few months of life, which may provide an immune priming benefit to other EPI vaccines.
Collapse
Affiliation(s)
- Agano Kiravu
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sophia Osawe
- Institute of Human Virology Nigeria, Abuja, Nigeria
| | - Anna-Ursula Happel
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Trishana Nundalall
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jerome Wendoh
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sophie Beer
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
| | - Nobomi Dontsa
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Olatogni Berenice Alinde
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Pam Datong
- Institute of Human Virology Nigeria, Abuja, Nigeria
| | - D William Cameron
- Divisions of Infectious Diseases and Respirology, University of Ottawa at the Ottawa Hospital, Ottawa, ON, Canada
| | - Kenneth Rosenthal
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Alash'le Abimiku
- Institute of Human Virology Nigeria, Abuja, Nigeria.,Institute of Human Virology, Department of Epidemiology and Prevention, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Heather B Jaspan
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Paediatrics and Global Health, University of Washington, Seattle, WA, United States
| | - Clive M Gray
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
15
|
Basu Roy R, Whittaker E, Seddon JA, Kampmann B. Tuberculosis susceptibility and protection in children. THE LANCET. INFECTIOUS DISEASES 2019; 19:e96-e108. [PMID: 30322790 PMCID: PMC6464092 DOI: 10.1016/s1473-3099(18)30157-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 01/27/2018] [Accepted: 02/09/2018] [Indexed: 12/14/2022]
Abstract
Children represent both a clinically important population susceptible to tuberculosis and a key group in whom to study intrinsic and vaccine-induced mechanisms of protection. After exposure to Mycobacterium tuberculosis, children aged under 5 years are at high risk of progressing first to tuberculosis infection, then to tuberculosis disease and possibly disseminated forms of tuberculosis, with accompanying high risks of morbidity and mortality. Children aged 5-10 years are somewhat protected, until risk increases again in adolescence. Furthermore, neonatal BCG programmes show the clearest proven benefit of vaccination against tuberculosis. Case-control comparisons from key cohorts, which recruited more than 15 000 children and adolescents in total, have identified that the ratio of monocytes to lymphocytes, activated CD4 T cell count, and a blood RNA signature could be correlates of risk for developing tuberculosis. Further studies of protected and susceptible populations are necessary to guide development of novel tuberculosis vaccines that could facilitate the achievement of WHO's goal to eliminate deaths from tuberculosis in childhood.
Collapse
Affiliation(s)
- Robindra Basu Roy
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, UK; Vaccines and Immunity Theme MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Elizabeth Whittaker
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, UK
| | - James A Seddon
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, UK
| | - Beate Kampmann
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, UK; Vaccines and Immunity Theme MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia.
| |
Collapse
|
16
|
Dzanibe S, Jaspan HB, Zulu MZ, Kiravu A, Gray CM. Impact of maternal HIV exposure, feeding status, and microbiome on infant cellular immunity. J Leukoc Biol 2019; 105:281-289. [PMID: 30577072 PMCID: PMC6923687 DOI: 10.1002/jlb.mr0318-120r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/17/2018] [Accepted: 12/05/2018] [Indexed: 01/02/2023] Open
Abstract
At least one-third of infants born in sub-Saharan Africa have been exposed to the effects of maternal HIV infection and antiretroviral treatment. Intrauterine HIV exposure is associated with increased rates of morbidity and mortality in children. Although the mechanisms responsible for poor infant health with HIV-1 exposure are likely to be multifactorial, we posit that the maternal environment during gestation and in the perinatal period results in altered infant immunity and is possibly the strongest contributing factor responsible for the disproportionally high infectious events among HIV-exposed infants who remain HIV uninfected. This review provides a synthesis of studies reporting the impact of intrauterine HIV exposure, feeding practices, and microbiota on immune ontogeny in the first year of life in HIV-exposed uninfected infants.
Collapse
Affiliation(s)
- Sonwabile Dzanibe
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Heather B. Jaspan
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Seattle Children’s Research Institute and Departments of Paediatrics and Global Health, University of Washington, Seattle, WA, USA
| | - Michael Z. Zulu
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Agano Kiravu
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Clive M. Gray
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
17
|
Yeganeh N, Watts DH, Xu J, Kerin T, Joao EC, Pilotto JH, Theron G, Gray G, Santos B, Fonseca R, Kreitchmann R, Pinto J, Mussi-Pinhata MM, Veloso V, Camarca M, Mofenson L, Moye J, Nielsen-Saines K. Infectious Morbidity, Mortality and Nutrition in HIV-exposed, Uninfected, Formula-fed Infants: Results From the HPTN 040/PACTG 1043 Trial. Pediatr Infect Dis J 2018; 37:1271-1278. [PMID: 29750766 PMCID: PMC6226320 DOI: 10.1097/inf.0000000000002082] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND HIV-exposed uninfected (HEU) infants are a growing population with potentially poor health outcomes. We evaluated morbidity and mortality in HEU formula-fed infants enrolled in the NICHD HPTN 040/PACTG 1043 trial. METHODS Infectious morbidity, mortality and undernutrition were evaluated within a cohort of 1000 HEU infants enrolled between April 2004 and April 2010 in Brazil (n = 766) and South Africa (n = 234) as part of the NICHD/HPTN 040 trial of 3 different antiretroviral regimens to decrease intrapartum HIV vertical transmission. RESULTS Twenty-three percent of infants had at least 1 infectious serious adverse effect. Infants born to mothers with <12 years of education [adjusted odds ratio (AOR), 2.6; 95% confidence interval [CI], 1.2-5.9), with maternal viral load of >1,000,000 copies/mL at delivery (AOR, 9.9; 95% CI, 1.6-63.1) were more likely to have infectious serious adverse effects. At 6 months, the infant mortality rate per 1000 live births overall was 22 ± 2.6, 9.1 ± 1.8 in Brazil and 64.1 ± 3 in South Africa. Undernutrition and stunting peaked at 1 month of age with 18% having a weight-for-age Z score ≤-2, and 22% with height for Z score ≤-2. The likelihood of infant mortality was greater among infants born in South Africa compared with Brazil (AOR, 6.2; 95% CI, 2.5-15.8), high maternal viral load (AOR, 1.7; 95% CI, 1.01-2.9) and birth weight-for-age Z score ≤-2 (AOR, 5.2; 95% CI, 1.8-14.8). CONCLUSIONS There were high rates of undernutrition, stunting and infectious serious adverse effect in this study's formula-fed HEU population. Suppressing maternal HIV viral load during the peripartum period may be a modifiable risk factor to decrease infant mortality.
Collapse
Affiliation(s)
- Nava Yeganeh
- David Geffen UCLA School of Medicine, Los Angeles, CA
| | - D. Heather Watts
- Office of the Global AIDS Coordinator and Health Diplomacy, U.S. Dept. of State, Washington D.C
| | | | - Tara Kerin
- David Geffen UCLA School of Medicine, Los Angeles, CA
| | - Esau C. Joao
- Hospital Federal dos Servidores do Estado, Rio de Janeiro, Brazil
| | - Jose Henrique Pilotto
- Hospital Geral de Nova Iguaçu, Nova Iguaçu and Laboratório de AIDS e Imunologia Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, (Fiocruz), Rio de Janeiro, Brazil, Perinatal HIV Research Unit
| | - Gerhard Theron
- Stellenbosch University/Tygerberg Hospital, Cape Town, South Africa
| | - Glenda Gray
- University of Witwatersrand/Chris Hani Baragwanath Hospital, Johannesburg, South Africa
| | | | | | | | - Jorge Pinto
- Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Valdilea Veloso
- Laboratório de Pesquisa Clínica em DST e AIDS - Instituto de Pesquisa Clínica Evandro Chagas - Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | | | - Lynne Mofenson
- Elisabeth Glaser Pediatric AIDS Foundation, Washington DC
| | - Jack Moye
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|
18
|
Falconer O, Newell ML, Jones CE. The Effect of Human Immunodeficiency Virus and Cytomegalovirus Infection on Infant Responses to Vaccines: A Review. Front Immunol 2018; 9:328. [PMID: 29552009 PMCID: PMC5840164 DOI: 10.3389/fimmu.2018.00328] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/06/2018] [Indexed: 12/11/2022] Open
Abstract
The success of prevention of mother to child transmission programs over the last two decades has led to an increasing number of infants who are exposed to human immunodeficiency virus (HIV), but who are not themselves infected (HIV-exposed, uninfected infants). Although the morbidity and mortality among HIV-exposed, uninfected infants is considerably lower than that among HIV-infected infants, they may remain at increased risk of infections in the first 2 years of life compared with their HIV-unexposed peers, especially in the absence of breastfeeding. There is some evidence of immunological differences in HIV-exposed, uninfected infants, which could play a role in susceptibility to infection. Cytomegalovirus (CMV) may contribute to the increased immune activation observed in HIV-exposed, uninfected infants. Infants born to HIV-infected women are at increased risk of congenital CMV infection, as well as early acquisition of postnatal CMV infection. In infants with HIV infection, CMV co-infection in early life is associated with higher morbidity and mortality. This review considers how HIV infection, HIV exposure, and CMV infection affect infant responses to vaccination, and explores possible immunological and other explanations for these findings. HIV-infected infants have lower vaccine-induced antibody concentrations following tetanus, diphtheria, pertussis, hepatitis B, and pneumococcal vaccination, although the clinical relevance of this difference is not known. Despite lower concentrations of maternal-specific antibody at birth, HIV-exposed, uninfected infants respond to vaccination at least as well as their HIV-unexposed uninfected peers. CMV infection leads to an increase in activation and differentiation of the whole T-cell population, but there is limited data on the effects of CMV infection on infant vaccine responses. In light of growing evidence of poor clinical outcomes associated with CMV infection in HIV-exposed, uninfected infants, further studies are particularly important in this group. A clearer understanding of the mechanisms by which maternal viral infections influence the developing infant immune system is critical to the success of maternal and infant vaccination strategies.
Collapse
Affiliation(s)
- Olivia Falconer
- Institute for Life Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Marie-Louise Newell
- Institute of Developmental Science, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Christine E Jones
- Institute for Life Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| |
Collapse
|
19
|
Tozetto-Mendoza TR, de Moraes Vasconcelos D, Ibrahim KY, Sartori AMC, Bezerra RC, de Freitas VLT, Shikanai-Yasuda MA. Role of T. cruzi exposure in the pattern of T cell cytokines among chronically infected HIV and Chagas disease patients. Clinics (Sao Paulo) 2017; 72:652-660. [PMID: 29236910 PMCID: PMC5706062 DOI: 10.6061/clinics/2017(11)02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/19/2017] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES The impact of Chagas disease (CD) in HIV-infected patients is relevant throughout the world. In fact, the characterization of the adaptive immune response in the context of co-infection is important for predicting the need for interventions in areas in which HIV and Chagas disease co-exist. METHODS We described and compared the frequency of cytokine-producing T cells stimulated with soluble antigen of Trypanosoma cruzi (T. cruzi) using a cytometric assay for the following groups: individuals with chronic Chagas disease (CHR, n=10), those with Chagas disease and HIV infection (CO, n=11), those with only HIV (HIV, n=14) and healthy individuals (C, n=15). RESULTS We found 1) a constitutively lower frequency of IL-2+ and IFN-γ+ T cells in the CHR group compared with the HIV, CO and healthy groups; 2) a suppressive activity of soluble T. cruzi antigen, which down-regulated IL-2+CD4+ and IFN-γ+CD4+ phenotypes, notably in the healthy group; 3) a down-regulation of inflammatory cytokines on CD8+ T cells in the indeterminate form of Chagas disease; and 4) a significant increase in IL-10+CD8+ cells distinguishing the indeterminate form from the cardiac/digestive form of Chagas disease, even in the presence of HIV infection. CONCLUSIONS Taken together, our data suggest the presence of an immunoregulatory response in chronic Chagas disease, which seems to be driven by T. cruzi antigens. Our findings provide new insights into immunotherapeutic strategies for people living with HIV/AIDS and Chagas disease.
Collapse
Affiliation(s)
- Tania Regina Tozetto-Mendoza
- Laboratorio de Imunologia (LIM 48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Laboratorio de Virologia (LIM 52), Universidade de Sao Paulo, Instituto de Medicina Tropical, Sao Paulo, SP, BR
- #These authors contributed equally to this work
| | - Dewton de Moraes Vasconcelos
- Laboratorio Dermatologia e Imunodeficiencias (LIM-56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- #These authors contributed equally to this work
| | - Karim Yaqub Ibrahim
- Divisao de Doencas Infecciosas e Parasitarias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Ana Marli Christovam Sartori
- Divisao de Doencas Infecciosas e Parasitarias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Rita C. Bezerra
- Laboratorio de Parasitologia (LIM 46), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Vera Lúcia Teixeira de Freitas
- Laboratorio de Imunologia (LIM 48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Departamento de Doencas Infecciosas e Parasitarias, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Maria Aparecida Shikanai-Yasuda
- Laboratorio de Imunologia (LIM 48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Departamento de Doencas Infecciosas e Parasitarias, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| |
Collapse
|
20
|
Garcia-Knight MA, Nduati E, Hassan AS, Nkumama I, Etyang TJ, Hajj NJ, Gambo F, Odera D, Berkley JA, Rowland-Jones SL, Urban B. Cytomegalovirus viraemia is associated with poor growth and T-cell activation with an increased burden in HIV-exposed uninfected infants. AIDS 2017; 31:1809-1818. [PMID: 28609400 PMCID: PMC5538302 DOI: 10.1097/qad.0000000000001568] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective: Factors associated with poor health in HIV-exposed-uninfected (HEU) infants are poorly defined. We describe the prevalence and correlates of cytomegalovirus (CMV) viraemia in HEU and HIV-unexposed-uninfected (HUU) infants, and quantify associations with anthropometric, haematological, and immunological outcomes. Design: Cross-sectional, including HEU and HUU infants from rural coastal Kenya. Methods: Infants aged 2–8 months were studied. The primary outcome was CMV viraemia and viral load, determined by quantitative PCR. Correlates were tested by logistic and linear regression; coefficients were used to describe associations between CMV viraemia and clinical/immunological parameters. Results: In total, 42 of 65 (64.6%) infants had CMV viraemia [median viral load, 3.0 (interquartile ranges: 2.7–3.5) log10 IU/ml]. Compared to community controls, HEU infants had six-fold increased odds of being viraemic (adjusted odds ratio 5.95 [95% confidence interval: 1.82–19.36], P = 0.003). Age, but not HEU/HUU status, was a strong correlate of CMV viral load (coefficient = −0.15, P = 0.009). CMV viral load associated negatively with weight-for-age (WAZ) Z-score (coefficient = −1.06, P = 0.008) and head circumference-for-age Z-score (coefficient = −1.47, P = 0.012) and positively with CD8+ T-cell coexpression of CD38/human leucocyte antigen DR (coefficient = 15.05, P = 0.003). Conclusion: The odds of having CMV viraemia was six-fold greater in HEU than HUU infants when adjusted for age. CMV viral load was associated with adverse growth and heightened CD8+ T-cell immune activation. Longitudinal assessments of the clinical effects of primary CMV infection and associated immunomodulation in early life in HEU and HUU populations are warranted.
Collapse
|
21
|
Smith C, Jalbert E, de Almeida V, Canniff J, Lenz LL, Mussi-Pinhata MM, Cohen RA, Yu Q, Amaral FR, Pinto J, Alarcon JO, Siberry G, Weinberg A. Altered Natural Killer Cell Function in HIV-Exposed Uninfected Infants. Front Immunol 2017; 8:470. [PMID: 28484464 PMCID: PMC5403425 DOI: 10.3389/fimmu.2017.00470] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 04/05/2017] [Indexed: 11/14/2022] Open
Abstract
Objectives HIV-exposed uninfected (HEU) infants have higher rates of severe and fatal infections compared with HIV-unexposed (HUU) infants, likely due to immune perturbations. We hypothesized that alterations in natural killer (NK) cell activity might occur in HEU infants and predispose them to severe infections. Design Case–control study using cryopreserved peripheral blood mononuclear cells (PBMCs) at birth and 6 months from HEU infants enrolled from 2002 to 2009 and HUU infants enrolled from 2011 to 2013. Methods NK cell phenotype and function were assessed by flow cytometry after 20-h incubation with and without K562 cells. Results The proportion of NK cells among PBMCs was lower at birth in 12 HEU vs. 22 HUU (1.68 vs. 10.30%, p < 0.0001) and at 6 months in 52 HEU vs. 72 HUU (3.09 vs. 4.65%, p = 0.0005). At birth, HEU NK cells demonstrated increased killing of K562 target cells (p < 0.0001) and increased expression of CD107a (21.65 vs. 12.70%, p = 0.047), but these differences resolved by 6 months. Stimulated HEU NK cells produced less interferon (IFN)γ at birth (0.77 vs. 2.64%, p = 0.008) and at 6 months (4.12 vs. 8.39%, p = 0.001), and showed reduced perforin staining at 6 months (66.95 vs. 77.30%, p = 0.0008). Analysis of cell culture supernatants indicated that lower NK cell activity in HEU was associated with reduced interleukin (IL)-12, IL-15, and IL-18. Addition of recombinant human IL-12 to stimulated HEU PBMCs restored IFNγ production to that seen in stimulated HUU cultures. Conclusion NK cell proportion, phenotype, and function are altered in HEU infants. NK cell cytotoxicity and degranulation are increased in HEU at birth, but HEU NK cells have reduced IFNγ and perforin production, suggesting an adequate initial response, but decreased functional reserve. NK cell function improved with addition of exogenous IL-12, implicating impaired production of IL-12 by accessory cells. Alterations in NK cell and accessory cell function may contribute to the increased susceptibility to infection in HEU infants.
Collapse
Affiliation(s)
- Christiana Smith
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| | - Emilie Jalbert
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| | - Volia de Almeida
- Federal University of Sao Carlos Biological and Health Sciences Center, Sao Carlos, Brazil
| | - Jennifer Canniff
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| | - Laurel L Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marisa M Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | | | - Fabiana R Amaral
- Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Jorge Pinto
- Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jorge O Alarcon
- Instituto de Medicina Tropical "Daniel A. Carrión" de la Universidad Nacional Mayor de San Marcos, Lima, Perú
| | - George Siberry
- Maternal and Pediatric Infectious Disease Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Adriana Weinberg
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
22
|
Njom Nlend AE, Nguwoh PS, Ngounouh CT, Tchidjou HK, Pieme CA, Otélé JM, Penlap V, Colizzi V, Moyou RS, Fokam J. HIV-Infected or -Exposed Children Exhibit Lower Immunogenicity to Hepatitis B Vaccine in Yaoundé, Cameroon: An Appeal for Revised Policies in Tropical Settings? PLoS One 2016; 11:e0161714. [PMID: 27656883 PMCID: PMC5033457 DOI: 10.1371/journal.pone.0161714] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022] Open
Abstract
Background Since 2005, anti-hepatitis B virus (anti-HBV) vaccine is part of the Expanded Program on Immunization (EPI) for infants born in Cameroon, with 99% anti-HBV coverage. In a context of generalized HIV epidemiology, we assessed paediatric anti-HBV vaccine response according to HIV status, feeding option and age in a tropical context. Methodology Prospective, observational and cross-sectional study conducted among 82 children (27 [IQR: 9–47] months, min-max: 6–59), after complete anti-HBV vaccination (Zilbrix Hepta: 10μg AgHBs) at the Essos Health Centre in Yaounde, Cameroon, classified as group-A: HIV unexposed (28), group-B: HIV-exposed/uninfected (29), group-C: HIV-infected (25). Quantitative anti-HBs ELISA was interpreted as “no”, “low-” or “protective-response” with <1, 1–10, or ≥10 IU/L respectively; with p-value<0.05 considered significant. Results Children were all HBV-unexposed (AcHBc-negative) and uninfected (HBsAg-negative). Response to anti-HBV vaccine was 80.49% (66/82), with only 45.12% (37/82) developed a protective-response (≥10IU/L). According to HIV status, 60.71% (17/28) developed a protective-response in group-A, vs. 51.72% (15/29) and 20% (5/25) in group-B and group-C respectively, Odds Ratio (OR): 2.627 [CI95% 0.933–7.500], p = 0.041. According to feeding option during first six months of life, 47.67% (21/45) developed a protective-response on exclusive breastfeeding vs. 43.24% (16/37) on mixed or formula feeding, OR: 1.148 [CI95% 0.437–3.026], p = 0.757. According to age, protective-response decreased significantly as children grow older: 58.33% (28/48) <24 months vs. 26.47% (9/34) ≥24 months, OR: 3.889 [CI95% 1.362–11.356], p = 0.004; and specifically 67.65% (23/34) ≤6 months vs. 0%, (0/5) 33–41 months, p = 0.008. Conclusions Anti-HBV vaccine provides low rate of protection (<50%) among children in general, and particularly if HIV-exposed, infected and/or older children. Implementing policies for early vaccination, specific immunization algorithm for HIV-exposed/infected children, and monitoring vaccine response would ensure effective protection in tropical settings, pending extensive/confirmatory investigations.
Collapse
Affiliation(s)
- Anne Esther Njom Nlend
- Pediatric Service, National Insurance Fund Welfare Hospital, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, Yaoundé, Cameroon
| | - Philippe Salomon Nguwoh
- Higher Institute of Health Professions, Yaoundé, Cameroon
- Ministry of Public Health, Yaoundé, Cameroon
- Institute of Science and Technology Applied to Health, Yaoundé, Cameroon
| | | | | | - Constant Anatole Pieme
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, Yaoundé, Cameroon
- Higher Institute of Health Professions, Yaoundé, Cameroon
| | | | - Véronique Penlap
- Higher Institute of Health Professions, Yaoundé, Cameroon
- Department of Biochemistry and Microbiology, Faculty of Science, University of Yaoundé 1, Yaoundé, Cameroon
| | - Vittorio Colizzi
- Chantal BIYA International Reference Centre for Research on HIV/AIDS prevention and management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
- UNESCO Interdisciplinary Board of Biotechnology, Rome, Italy
| | - Roger Somo Moyou
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, Yaoundé, Cameroon
- Chantal BIYA International Reference Centre for Research on HIV/AIDS prevention and management, Yaoundé, Cameroon
- Institute of Research in Medicine and Medicinal plants, Yaoundé, Cameroon
| | - Joseph Fokam
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, Yaoundé, Cameroon
- Chantal BIYA International Reference Centre for Research on HIV/AIDS prevention and management, Yaoundé, Cameroon
- University of Rome Tor Vergata, Rome, Italy
- * E-mail:
| |
Collapse
|
23
|
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: 1.8] [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
|
24
|
Ruck C, Reikie BA, Marchant A, Kollmann TR, Kakkar F. Linking Susceptibility to Infectious Diseases to Immune System Abnormalities among HIV-Exposed Uninfected Infants. Front Immunol 2016; 7:310. [PMID: 27594857 PMCID: PMC4990535 DOI: 10.3389/fimmu.2016.00310] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/02/2016] [Indexed: 12/22/2022] Open
Abstract
HIV-exposed uninfected (HEU) infants experience increased overall mortality from infectious causes when compared to HIV-unexposed uninfected (HU) infants. This is the case in both the resource-rich and resource-limited settings. Here, we explore the concept that specific types of infectious diseases that are more common among HEU infants could provide clues as to the potential underlying immunological abnormalities. The most commonly reported infections in HEU vs. HU infants are caused by encapsulated bacteria, suggesting the existence of a less effective humoral (antibody, complement) immune response. Decreased transplacental transfer of protective maternal antibodies has consistently been observed among HEU newborns, suggesting that this may indeed be one of the key drivers of their susceptibility to infections with encapsulated bacteria. Reassuringly, HEU humoral response to vaccination appears to be well conserved. While there appears to be an increase in overall incidence of acute viral infections, no specific pattern of acute viral infections has emerged; and although there is evidence of increased chronic viral infection from perinatal transmission of hepatitis C and cytomegalovirus, no data exist to suggest an increase in adverse outcomes. Thus, no firm conclusions about antiviral effector mechanisms can be drawn. However, the most unusual of reported infections among the HEU have been opportunistic infections, suggesting the possibility of underlying defects in CD4 helper T cells and overall immune regulatory function. This may relate to the observation that the immunological profile of HEUs indicates a more activated T cell profile as well as a more inflammatory innate immune response. However, both of these observations appear transient, marked in early infancy, but no longer evident later in life. The causes of these early-life changes in immune profiles are likely multifactorial and may be related to in utero exposure to HIV, but also to increased environmental exposure to pathogens from sicker household contacts, in utero and postnatal antiretroviral drug exposure, and, in certain circumstances, differences in mode of feeding. The relative importance of each of these factors will be important to delineate in an attempt to identify those HEU at highest risk of adverse outcomes for targeted interventions.
Collapse
Affiliation(s)
- Candice Ruck
- Department of Pediatrics, BC Women's and Children's Hospital, University of British Columbia , Vancouver, BC , Canada
| | - Brian A Reikie
- Department of Surgery, University of Manitoba , Winnipeg, MB , Canada
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles , Charleroi , Belgium
| | - Tobias R Kollmann
- Department of Pediatrics, BC Women's and Children's Hospital, University of British Columbia , Vancouver, BC , Canada
| | - Fatima Kakkar
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal , Montréal, QC , Canada
| |
Collapse
|
25
|
Evans C, Jones CE, Prendergast AJ. HIV-exposed, uninfected infants: new global challenges in the era of paediatric HIV elimination. THE LANCET. INFECTIOUS DISEASES 2016; 16:e92-e107. [PMID: 27049574 DOI: 10.1016/s1473-3099(16)00055-4] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 01/01/2016] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
The number of infants infected with HIV is declining with the rise in interventions for the elimination of paediatric HIV infection, but the number of uninfected infants exposed to HIV through their HIV-infected mothers is increasing. Interest in the health outcomes of HIV-exposed, uninfected infants has grown in the past decade, with several studies suggesting that these infants have increased mortality rates, increased infectious morbidity, and impaired growth compared with HIV-unexposed infants. However, heterogeneous results might reflect the inherent challenges in studies of HIV-exposed, uninfected infants, which need large populations with appropriate, contemporaneous comparison groups and repeated HIV testing throughout the period of breastfeeding. We review the effects of HIV exposure on mortality, morbidity, and growth, discuss the immunological abnormalities identified so far, and provide an overview of interventions that could be effective in this susceptible population. As the number of infants infected with HIV declines, the health needs of HIV-exposed, uninfected infants should be prioritised further, to ensure that post-2015 Sustainable Development Goals are achieved.
Collapse
Affiliation(s)
- Ceri Evans
- Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Christine E Jones
- Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|