1
|
Yin L, Venturi GM, Barfield R, Fischer BM, Kim-Chang JJ, Chan C, De Paris K, Goodenow MM, Sleasman JW. Maternal immunity shapes biomarkers of germinal center development in HIV-exposed uninfected infants. Front Immunol 2024; 15:1443886. [PMID: 39328414 PMCID: PMC11424517 DOI: 10.3389/fimmu.2024.1443886] [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: 06/04/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Introduction HIV-exposed uninfected (HEU) infants exhibit elevated pro-inflammatory biomarkers that persist after birth. However, comprehensive assessments of bioprofiles associated with immune regulation and development in pregnant women with HIV (PWH) and HEU infants has not been performed. Maternal immunity in PWH may be imprinted on their HEU newborns, altering immune bioprofiles during early immune development. Methods Cryopreserved paired plasma samples from 46 HEU infants and their mothers enrolled in PACTG 316, a clinical trial to prevent perinatal HIV-1 transmission were analyzed. PWH received antiretrovirals (ARV) and had either fully suppressed or unsuppressed viral replication. Maternal blood samples obtained during labor and infant samples at birth and 6 months were measured for 21 biomarkers associated with germinal centers (GC), macrophage activation, T-cell activation, interferon gamma (IFN-γ)-inducible chemokines, and immune regulatory cytokines using Mesoscale assays. Pregnant women without HIV (PWOH) and their HIV unexposed uninfected (HUU) newborns and non-pregnant women without HIV (NPWOH) served as reference groups. Linear regression analysis fitted for comparison among groups and adjusted for covariant(s) along with principal component analysis performed to assess differences among groups. Results Compared with NPWOH, PWOH displayed higher levels of GC, macrophage, and regulatory biomarkers. PWH compared to PWOH displayed elevated GC, T cell activation, and IFN-γ-inducible chemokines biomarkers at delivery. Similar to their mothers, HEU infants had elevated GC, macrophage, and IFN-γ-inducible chemokines, as well as elevated anti-inflammatory cytokines, IL-10 and IL-1RA. Across all mother/newborn dyads, multiple biomarkers positively correlated, providing further evidence that maternal inflammation imprints on newborn bioprofiles. By 6 months, many HEU biomarkers normalized to levels similar to HUU infants, but some GC and inflammatory biomarkers remained perturbed. Bioprofiles in PWH and HEU infants were similar regardless of the extent of maternal viral suppression by ARV. Conclusions GC immune pathways are perturbed in HEU newborns, but immune regulatory responses down regulate inflammation during early infancy, indicating a transient inflammatory effect. However, several GC biomarkers that may alter immune development remain perturbed.
Collapse
Affiliation(s)
- Li Yin
- Molecular HIV Host Interactions Section, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Guglielmo M. Venturi
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Bernard M. Fischer
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Julie J. Kim-Chang
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Institute of Global Health and Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Maureen M. Goodenow
- Molecular HIV Host Interactions Section, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - John W. Sleasman
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
2
|
Sanders AFP, Tirado B, Seider NA, Triplett RL, Lean RE, Neil JJ, Miller JP, Tillman R, Smyser TA, Barch DM, Luby JL, Rogers CE, Smyser CD, Warner BB, Chen E, Miller GE. Prenatal exposure to maternal disadvantage-related inflammatory biomarkers: associations with neonatal white matter microstructure. Transl Psychiatry 2024; 14:72. [PMID: 38307841 PMCID: PMC10837200 DOI: 10.1038/s41398-024-02782-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/04/2024] Open
Abstract
Prenatal exposure to heightened maternal inflammation has been associated with adverse neurodevelopmental outcomes, including atypical brain maturation and psychiatric illness. In mothers experiencing socioeconomic disadvantage, immune activation can be a product of the chronic stress inherent to such environmental hardship. While growing preclinical and clinical evidence has shown links between altered neonatal brain development and increased inflammatory states in utero, the potential mechanism by which socioeconomic disadvantage differentially impacts neural-immune crosstalk remains unclear. In the current study, we investigated associations between socioeconomic disadvantage, gestational inflammation, and neonatal white matter microstructure in 320 mother-infant dyads over-sampled for poverty. We analyzed maternal serum levels of four cytokines (IL-6, IL-8, IL-10, TNF-α) over the course of pregnancy in relation to offspring white matter microstructure and socioeconomic disadvantage. Higher average maternal IL-6 was associated with very low socioeconomic status (SES; INR < 200% poverty line) and lower neonatal corticospinal fractional anisotropy (FA) and lower uncinate axial diffusivity (AD). No other cytokine was associated with SES. Higher average maternal IL-10 was associated with lower FA and higher radial diffusivity (RD) in corpus callosum and corticospinal tracts, higher optic radiation RD, lower uncinate AD, and lower FA in inferior fronto-occipital fasciculus and anterior limb of internal capsule tracts. SES moderated the relationship between average maternal TNF-α levels during gestation and neonatal white matter diffusivity. When these interactions were decomposed, the patterns indicated that this association was significant and positive among very low SES neonates, whereby TNF-α was inversely and significantly associated with inferior cingulum AD. By contrast, among the more advantaged neonates (lower-to-higher SES [INR ≥ 200% poverty line]), TNF-α was positively and significantly associated with superior cingulum AD. Taken together, these findings suggest that the relationship between prenatal cytokine exposure and white matter microstructure differs as a function of SES. These patterns are consistent with a scenario where gestational inflammation's effects on white matter development diverge depending on the availability of foundational resources in utero.
Collapse
Affiliation(s)
- Ashley F P Sanders
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Brian Tirado
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nicole A Seider
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Regina L Triplett
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rachel E Lean
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jeffrey J Neil
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - J Philip Miller
- Division of Biostatistics, Institute for Informatics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rebecca Tillman
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tara A Smyser
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Deanna M Barch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Psychological and Brain Sciences, Washington University School of Medicine, St. Louis, MO, 63130, USA
| | - Joan L Luby
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Cynthia E Rogers
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Christopher D Smyser
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Newborn Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Edith Chen
- Institute for Policy Research, Northwestern University, Evanston, IL, 60208, USA
- Department of Psychology, Northwestern University, Evanston, IL, 60208, USA
| | - Gregory E Miller
- Institute for Policy Research, Northwestern University, Evanston, IL, 60208, USA
- Department of Psychology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
3
|
Martinez VO, Dos Santos NR, Bah HAF, Junior EAG, Costa DO, Menezes-Filho JA. Impact of chronic toxoplasmosis in pregnancy: association between maternal seropositivity for Toxoplasma gondii IgG antibodies and fetal growth restriction. Parasitol Res 2023; 123:25. [PMID: 38072839 DOI: 10.1007/s00436-023-08068-y] [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: 08/18/2023] [Accepted: 10/20/2023] [Indexed: 12/18/2023]
Abstract
Insults caused by acute infections during the gestational period on fetal development are known; however, new evidence suggests that chronic infectious diseases can also impact the maternal immune status and lead to negative consequences for the neonate. This study investigated the association between the prevalence of specific antibodies in pregnant women and alterations in fetal development at birth. A follow-up study evaluated women during the gestational period and their respective newborns at delivery time. The pregnant women were tested for the presence of antibodies to infectious agents: Toxoplasma gondii (T. gondii), cytomegalovirus (CMV), syphilis, human immunodeficiency virus (HIV), hepatitis B and C. Semi-structured questionnaires were administered to the pregnant women at the time of recruitment after obtaining informed consent. Detailed information about the newborns was extracted from medical records. The seroprevalence of chronic T. gondii infection, as determined by the presence of IgG antibodies against the protozoan, was found to be 56.2%, while the overall prevalence of CMV IgG antibodies was 96.3%. Non-primiparous pregnant women from socio-economic classes, less affluent groups, and skilled working-class individuals had higher chances of testing positive for specific T. gondii IgG antibodies. Newborns classified as small for gestational age represented 12.9% of the total. Those born to mothers seropositive for anti-T. gondii IgG antibodies were 9.4 times more likely to be born small for gestational age (p = 0.035). The results suggest that chronic T. gondii infection may contribute to higher rates of newborns with growth restriction. These findings add to a growing body of evidence regarding the impact of chronic infectious diseases on intrauterine fetal development.
Collapse
Affiliation(s)
- Victor Otero Martinez
- Graduate Program in Pharmacy, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil.
| | - Nathália Ribeiro Dos Santos
- Graduate Program in Pharmacy, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
- Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Homègnon Antonin Ferréol Bah
- Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
- Graduate Program in Public Health, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | - Daisy Oliveira Costa
- Graduate Program in Pharmacy, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
- Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - José Antonio Menezes-Filho
- Graduate Program in Pharmacy, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
- Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
- Graduate Program in Public Health, Federal University of Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
4
|
Dubois V, Chatagnon J, Depessemier M, Locht C. Maternal acellular pertussis vaccination in mice impairs cellular immunity to Bordetella pertussis infection in offspring. JCI Insight 2023; 8:e167210. [PMID: 37581930 PMCID: PMC10561720 DOI: 10.1172/jci.insight.167210] [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: 11/15/2022] [Accepted: 08/08/2023] [Indexed: 08/17/2023] Open
Abstract
Given the resurgence of pertussis, several countries have introduced maternal tetanus, diphtheria, and acellular pertussis (aP) vaccination during pregnancy to protect young infants against severe pertussis. Although protective against the disease, the effect of maternal aP vaccination on bacterial colonization of the offspring is unknown. Here, we used a mouse model to demonstrate that maternal aP immunization, either before or during pregnancy, protects pups from lung colonization by Bordetella pertussis. However, maternal aP vaccination resulted in significantly prolonged nasal carriage of B. pertussis by inhibiting the natural recruitment of IL-17-producing resident memory T cells and ensuing neutrophil influx in the nasal tissue, especially of those with proinflammatory and cytotoxic properties. Prolonged nasal carriage after aP vaccination is due to IL-4 signaling, as prolonged nasal carriage is abolished in IL-4Rα-/- mice. The effect of maternal aP vaccination can be transferred transplacentally to the offspring or via breastfeeding and is long-lasting, as it persists into adulthood. Maternal aP vaccination may, thus, augment the B. pertussis reservoir.
Collapse
|
5
|
Gonçalves J, Melro M, Alenquer M, Araújo C, Castro-Neves J, Amaral-Silva D, Ferreira F, Ramalho JS, Charepe N, Serrano F, Pontinha C, Amorim MJ, Soares H. Balance between maternal antiviral response and placental transfer of protection in gestational SARS-CoV-2 infection. JCI Insight 2023; 8:e167140. [PMID: 37490342 PMCID: PMC10544212 DOI: 10.1172/jci.insight.167140] [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: 11/11/2022] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
The intricate interplay between maternal immune response to SARS-CoV-2 and the transfer of protective factors to the fetus remains unclear. By analyzing mother-neonate dyads from second and third trimester SARS-CoV-2 infections, our study shows that neutralizing antibodies (NAbs) are infrequently detected in cord blood. We uncovered that this is due to impaired IgG-NAb placental transfer in symptomatic infection and to the predominance of maternal SARS-CoV-2 NAbs of the IgA and IgM isotypes, which are prevented from crossing the placenta. Crucially, the balance between maternal antiviral response and transplacental transfer of IgG-NAbs appears to hinge on IL-6 and IL-10 produced in response to SARS-CoV-2 infection. In addition, asymptomatic maternal infection was associated with expansion of anti-SARS-CoV-2 IgM and NK cell frequency. Our findings identify a protective role for IgA/IgM-NAbs in gestational SARS-CoV-2 infection and open the possibility that the maternal immune response to SARS-CoV-2 infection might benefit the neonate in 2 ways, first by skewing maternal immune response toward immediate viral clearance, and second by endowing the neonate with protective mechanisms to curtail horizontal viral transmission in the critical postnatal period, via the priming of IgA/IgM-NAbs to be transferred by the breast milk and via NK cell expansion in the neonate.
Collapse
Affiliation(s)
- Juliana Gonçalves
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Magda Melro
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Marta Alenquer
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | - Catarina Araújo
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Júlia Castro-Neves
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Daniela Amaral-Silva
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Filipe Ferreira
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | | | - Nádia Charepe
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Fátima Serrano
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Carlos Pontinha
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | - Helena Soares
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| |
Collapse
|
6
|
Ibrahim A, Warton FL, Fry S, Cotton MF, Jacobson SW, Jacobson JL, Molteno CD, Little F, van der Kouwe AJW, Laughton B, Meintjes EM, Holmes MJ. Maternal ART throughout gestation prevents caudate volume reductions in neonates who are HIV exposed but uninfected. Front Neurosci 2023; 17:1085589. [PMID: 36968507 PMCID: PMC10035579 DOI: 10.3389/fnins.2023.1085589] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/25/2023] [Indexed: 03/12/2023] Open
Abstract
IntroductionSuccessful programmes for prevention of vertical HIV transmission have reduced the risk of infant HIV infection in South Africa from 8% in 2008 to below 1% in 2018/2019, resulting in an increasing population of children exposed to HIV perinatally but who are uninfected (HEU). However, the long-term effects of HIV and antiretroviral treatment (ART) exposure on the developing brain are not well understood. Whereas children who are HEU perform better than their HIV-infected counterparts, they demonstrate greater neurodevelopmental delay than children who are HIV unexposed and uninfected (HUU), especially in resource-poor settings. Here we investigate subcortical volumetric differences related to HIV and ART exposure in neonates.MethodsWe included 120 infants (59 girls; 79 HEU) born to healthy women with and without HIV infection in Cape Town, South Africa, where HIV sero-prevalence approaches 30%. Of the 79 HEU infants, 40 were exposed to ART throughout gestation (i.e., mothers initiated ART pre conception; HEU-pre), and 39 were exposed to ART for part of gestation (i.e., mothers initiated ART post conception; HEU-post). Post-conception mothers had a mean (± SD) gestational age (GA) of 15.4 (± 5.7) weeks at ART initiation. Mothers with HIV received standard care fixed drug combination ART (Tenofovir/Efavirenz/Emtricitabine). Infants were imaged unsedated on a 3T Skyra (Siemens, Erlangen, Germany) at mean GA equivalent of 41.5 (± 1.0) weeks. Selected regions (caudate, putamen, pallidum, thalamus, cerebellar hemispheres and vermis, and corpus callosum) were manually traced on T1-weighted images using Freeview.ResultsHEU neonates had smaller left putamen volumes than HUU [β (SE) = −90.3 (45.3), p = 0.05] and caudate volume reductions that depended on ART exposure duration in utero. While the HEU-pre group demonstrated no caudate volume reductions compared to HUU, the HEU-post group had smaller caudate volumes bilaterally [β (SE) = −145.5 (45.1), p = 0.002, and −135.7 (49.7), p = 0.008 for left and right caudate, respectively].DiscussionThese findings from the first postnatal month suggest that maternal ART throughout gestation is protective to the caudate nuclei. In contrast, left putamens were smaller across all HEU newborns, despite maternal ART.
Collapse
Affiliation(s)
- Abdulmumin Ibrahim
- Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, Biomedical Engineering Research Centre, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Fleur L. Warton
- Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, Biomedical Engineering Research Centre, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- *Correspondence: Fleur L. Warton,
| | - Samantha Fry
- Department of Paediatrics and Child Health and Tygerberg Children’s Hospital, Faculty of Medicine and Health Sciences, Family Centre for Research with Ubuntu, Stellenbosch University, Stellenbosch, South Africa
| | - Mark F. Cotton
- Department of Paediatrics and Child Health and Tygerberg Children’s Hospital, Faculty of Medicine and Health Sciences, Family Centre for Research with Ubuntu, Stellenbosch University, Stellenbosch, South Africa
| | - Sandra W. Jacobson
- Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, Biomedical Engineering Research Centre, University of Cape Town, Cape Town, South Africa
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Joseph L. Jacobson
- Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, Biomedical Engineering Research Centre, University of Cape Town, Cape Town, South Africa
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Christopher D. Molteno
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Francesca Little
- Department of Statistical Sciences, University of Cape Town, Cape Town, South Africa
| | - Andre J. W. van der Kouwe
- Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, Biomedical Engineering Research Centre, University of Cape Town, Cape Town, South Africa
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
- Department of Radiology, Harvard Medical School, Boston, MA, United States
| | - Barbara Laughton
- Department of Paediatrics and Child Health and Tygerberg Children’s Hospital, Faculty of Medicine and Health Sciences, Family Centre for Research with Ubuntu, Stellenbosch University, Stellenbosch, South Africa
| | - Ernesta M. Meintjes
- Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, Biomedical Engineering Research Centre, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Cape Universities Body Imaging Centre, University of Cape Town, Cape Town, South Africa
- Ernesta M. Meintjes,
| | - Martha J. Holmes
- Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, Biomedical Engineering Research Centre, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
7
|
Jordan-Paiz A, Martrus G, Steinert FL, Kaufmann M, Sagebiel AF, Schreurs RRCE, Rechtien A, Baumdick ME, Jung JM, Möller KJ, Wegner L, Grüttner C, Richert L, Thünauer R, Schroeder-Schwarz J, van Goudoever JB, Geijtenbeek TBH, Altfeld M, Pals ST, Perez D, Klarenbeek PL, Tomuschat C, Sauter G, Königs I, Schumacher U, Friese MA, Melling N, Reinshagen K, Bunders MJ. CXCR5 +PD-1 ++ CD4 + T cells colonize infant intestines early in life and promote B cell maturation. Cell Mol Immunol 2023; 20:201-213. [PMID: 36600048 PMCID: PMC9886971 DOI: 10.1038/s41423-022-00944-4] [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: 01/18/2022] [Accepted: 10/26/2022] [Indexed: 01/06/2023] Open
Abstract
Gastrointestinal infections are a major cause for serious clinical complications in infants. The induction of antibody responses by B cells is critical for protective immunity against infections and requires CXCR5+PD-1++ CD4+ T cells (TFH cells). We investigated the ontogeny of CXCR5+PD-1++ CD4+ T cells in human intestines. While CXCR5+PD-1++ CD4+ T cells were absent in fetal intestines, CXCR5+PD-1++ CD4+ T cells increased after birth and were abundant in infant intestines, resulting in significant higher numbers compared to adults. These findings were supported by scRNAseq analyses, showing increased frequencies of CD4+ T cells with a TFH gene signature in infant intestines compared to blood. Co-cultures of autologous infant intestinal CXCR5+PD-1+/-CD4+ T cells with B cells further demonstrated that infant intestinal TFH cells were able to effectively promote class switching and antibody production by B cells. Taken together, we demonstrate that functional TFH cells are numerous in infant intestines, making them a promising target for oral pediatric vaccine strategies.
Collapse
Affiliation(s)
- Ana Jordan-Paiz
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Glòria Martrus
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Fenja L Steinert
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Max Kaufmann
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Adrian F Sagebiel
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Renée R C E Schreurs
- Department of Experimental Immunology; Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Anne Rechtien
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
- Partner Site Hamburg-Lübeck-Borstel-Riems, German Center for Infection Research (DZIF), Hamburg, 20246, Germany
| | - Martin E Baumdick
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Johannes M Jung
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Kimberly J Möller
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Lucy Wegner
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Cordula Grüttner
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Laura Richert
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale, Bordeaux Population Health Research Center UMR1219 and INRIA SISTM Team, Bordeaux, 33000, France
| | - Roland Thünauer
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Jennifer Schroeder-Schwarz
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Johannes B van Goudoever
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology; Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Marcus Altfeld
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Paul L Klarenbeek
- Department of Rheumatology and Clinical Immunology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1007 MB, The Netherlands
- Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Christian Tomuschat
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Ingo Königs
- Department of Pediatric Surgery, Altona Children's Hospital, Hamburg, 22763, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Madeleine J Bunders
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany.
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.
| |
Collapse
|
8
|
Jalbert E, Ghosh T, Smith C, Amaral FR, Mussi-Pinhata MM, Weinberg A. Impaired functionality of antigen presenting cells in HIV- exposed uninfected infants in the first six months of life. Front Immunol 2022; 13:960313. [PMID: 36032106 PMCID: PMC9411519 DOI: 10.3389/fimmu.2022.960313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
HIV-exposed uninfected infants (HEU) have increased morbidity and mortality due to infections in the first 6 months of life that tapers down to 2 years of life. The underlying immunologic defects remain undefined. We investigated antigen-presenting cells (APC) by comparing the phenotype of unstimulated APC, responses to toll-like receptor (TLR) stimulation, and ability to activate natural killer (NK) cells in 24 HEU and 64 HIV-unexposed infants (HUU) at 1-2 days of life (birth) and 28 HEU and 45 HUU at 6 months of life. At birth, unstimulated APC showed higher levels of activation and cytokine production in HEU than HUU and stimulation with TLR agonists revealed lower expression of inflammatory cytokines and activation markers, but similar expression of IL10 regulatory cytokine, in APC from HEU compared to HUU. Differences were still present at 6 months of life. From birth to 6 months, APC underwent extensive phenotypic and functional changes in HUU and minimal changes in HEU. TLR stimulation also generated lower NK cell expression of CD69 and/or IFNγ in HEU compared with HUU at birth and 6 months. In vitro experiments showed that NK IFNγ expression depended on APC cytokine secretion in response to TLR stimulation. Ex vivo IL10 supplementation decreased APC-mediated NK cell activation measured by IFNγ expression. We conclude that APC maturation was stunted or delayed in the first 6 months of life in HEU compared with HUU. Deficient inflammatory APC responses and/or the imbalance between inflammatory and regulatory responses in HEU may play an important role in their increased susceptibility to severe infections.
Collapse
Affiliation(s)
- Emilie Jalbert
- Department of Pediatrics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Tusharkanti Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Christiana Smith
- Department of Pediatrics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Fabiana R. Amaral
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marisa M. Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Adriana Weinberg
- Department of Pediatrics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine and Pathology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: Adriana Weinberg,
| |
Collapse
|
9
|
Understanding the Immune System in Fetal Protection and Maternal Infections during Pregnancy. J Immunol Res 2022; 2022:7567708. [PMID: 35785037 PMCID: PMC9249541 DOI: 10.1155/2022/7567708] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/20/2022] [Indexed: 11/18/2022] Open
Abstract
The fetal-maternal immune system determines the fate of pregnancy. The trophoblast cells not only give an active response against external stimuli but are also involved in secreting most of the cytokines. These cells have an essential function in fetal acceptance or fetal rejection. Other immune cells also play a pivotal role in carrying out a successful pregnancy. The disruption in this mechanism may lead to harmful effects on pregnancy. The placenta serves as an immune barrier in fetus protection against invading pathogens. Once the infections prevail, they may localize in placental and fetal tissues, and the presence of inflammation due to cytokines may have detrimental effects on pregnancy. Moreover, some pathogens are responsible for congenital fetal anomalies and affect almost all organs of the developing fetus. This review article is designed to address the bacterial and viral infections that threaten pregnancy and their possible outcomes. Moreover, training of the fetal immune system against the exposure of infections and the role of CD49a + NK cells in embryonic development will also be highlighted.
Collapse
|
10
|
Teunissen CE, Rohlwink U, Pajkrt D, Naudé PJW. Biomarkers of Tuberculous Meningitis and Pediatric Human Immunodeficiency Virus on the African Continent. Front Neurol 2022; 13:793080. [PMID: 35665032 PMCID: PMC9160376 DOI: 10.3389/fneur.2022.793080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Biomarkers in body fluids are helpful objective tools in diagnosis, prognosis and monitoring of (therapeutic) responses of many neurological diseases. Cerebrospinal fluid (CSF) biomarkers are part of the diagnostic toolbox for infectious neurological diseases. Tuberculous meningitis (TBM) and Human immunodeficiency virus (HIV), are important burdens of disease in Africa and can negatively affect brain health. Two thirds of the world's population of people living with HIV reside in sub-Saharan Africa and 25% of the global burden of tuberculosis (TB) is carried by the African continent. Neuroinflammation and damage of specific neuronal cell types are key constituents in the pathophysiology of these central nervous system (CNS) diseases, and important potential sources of circulating biomarkers. In this review, we summarize current research in the use of biomarkers in TBM and pediatric HIV as case demonstrations for high prevalence neurological diseases in Africa. Inflammatory molecules, primarily when detected in CSF, appear to have diagnostic value in these diseases, especially when measured as profiles. Brain injury molecules, such as S100, Neuron specific enolase and glial fibrillary acidic protein may have prognostic value in TBM, but more studies are needed. There is a need for more cost-economic and high sensitivity technologies to drive further biomarker discoveries and translate into healthcare improvements for these important healthcare problems in a globally fair way.
Collapse
Affiliation(s)
- Charlotte Elisabeth Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Ursula Rohlwink
- Division of Neurosurgery, Neuroscience Institute, Department of Surgery, University of Cape Town, Cape Town, South Africa
- The Francis Crick Institute, London, United Kingdom
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, Netherlands
| | - Petrus J. W. Naudé
- Department of Psychiatry and Mental Health, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
11
|
Li Q, Yang S, Zhang X, Liu X, Wu Z, Qi Y, Guan W, Ren M, Zhang S. Maternal Nutrition During Late Gestation and Lactation: Association With Immunity and the Inflammatory Response in the Offspring. Front Immunol 2022; 12:758525. [PMID: 35126349 PMCID: PMC8814630 DOI: 10.3389/fimmu.2021.758525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
The immature immune system at birth and environmental stress increase the risk of infection in nursing pigs. Severe infection subsequently induces intestinal and respiratory diseases and even cause death of pigs. The nutritional and physiological conditions of sows directly affect the growth, development and disease resistance of the fetus and newborn. Many studies have shown that providing sows with nutrients such as functional oligosaccharides, oils, antioxidants, and trace elements could regulate immunity and the inflammatory response of piglets. Here, we reviewed the positive effects of certain nutrients on milk quality, immunoglobulin inflammatory response, oxidative stress, and intestinal microflora of sows, and further discuss the effects of these nutrients on immunity and the inflammatory response in the offspring.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoli Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xinghong Liu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhihui Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yingao Qi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Man Ren
- College of Animal Science, Anhui Science and Technology University, Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, Fengyang, China
- *Correspondence: Man Ren, ; Shihai Zhang,
| | - Shihai Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
- *Correspondence: Man Ren, ; Shihai Zhang,
| |
Collapse
|
12
|
Immunopathogenesis in HIV-associated pediatric tuberculosis. Pediatr Res 2022; 91:21-26. [PMID: 33731810 PMCID: PMC8446109 DOI: 10.1038/s41390-021-01393-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/25/2020] [Accepted: 01/18/2021] [Indexed: 11/09/2022]
Abstract
Tuberculosis (TB) is an increasing global emergency in human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS) patients, in which host immunity is dysregulated and compromised. However, the pathogenesis and efficacy of therapeutic strategies in HIV-associated TB in developing infants are essentially lacking. Bacillus Calmette-Guerin vaccine, an attenuated live strain of Mycobacterium bovis, is not adequately effective, which confers partial protection against Mycobacterium tuberculosis (Mtb) in infants when administered at birth. However, pediatric HIV infection is most devastating in the disease progression of TB. It remains challenging whether early antiretroviral therapy (ART) could maintain immune development and function, and restore Mtb-specific immune function in HIV-associated TB in children. A better understanding of the immunopathogenesis in HIV-associated pediatric Mtb infection is essential to provide more effective interventions, reducing the risk of morbidity and mortality in HIV-associated Mtb infection in infants. IMPACT: Children living with HIV are more likely prone to opportunistic infection, predisposing high risk of TB diseases. HIV and Mtb coinfection in infants may synergistically accelerate disease progression. Early ART may probably induce immune reconstitution inflammatory syndrome and TB pathology in HIV/Mtb coinfected infants.
Collapse
|
13
|
Takahashi M, Makino S, Iizuka H, Noguchi M, Yoshida K. Chronic active Epstein-Barr virus-associated secondary hemophagocytic lymphohistiocytosis in pregnancy: a case report. BMC Pregnancy Childbirth 2021; 21:681. [PMID: 34620104 PMCID: PMC8495184 DOI: 10.1186/s12884-021-04150-4] [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: 07/28/2021] [Accepted: 09/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Secondary hemophagocytic lymphohistiocytosis (sHLH) is a rare and fatal disease characterized by uncontrolled immune cell activation that can lead to a cytokine storm. Unfortunately, this condition can occur even during pregnancy, threatening both maternal and fetal lives. CASE PRESENTATION A 23-year-old nulliparous woman at 26 weeks of gestation presented with continuous fever, coughing, and sore throat. Upon arrival at our hospital, her temperature was >38°C and laboratory findings indicated cytopenia (neutrophil count, 779/μL; hemoglobin level, 10.2 g/dL; platelet count, 29,000/μL), elevated ferritin level (1,308 ng/mL), and elevated soluble interleukin-2 receptor level (11,200 U/mL). Computed tomography showed marked splenomegaly. Bone marrow examination revealed hemophagocytosis, and blood examination showed a plasma Epstein-Barr virus (EBV) DNA level of 8.9 × 105 copies/μg. The monoclonal proliferation of EBV-infected T cells was confirmed by Southern blotting, and the patient was diagnosed with chronic active EBV-associated sHLH and T-cell lymphoproliferative disease. Immediately after admission, the patient's condition suddenly deteriorated. She developed shock and disseminated intravascular coagulation, requiring endotracheal intubation along with methylprednisolone pulse and etoposide therapy. Although the patient recovered, she delivered a stillborn baby. After delivery, she was treated with reduced-dose dexamethasone, etoposide, ifosfamide, and carboplatin (DeVIC) and steroid (dexamethasone), methotrexate, ifosfamide, L-asparaginase, and etoposide (SMILE) chemotherapies. Five months after diagnosis, she received human leukocyte antigen-haploidentical allogeneic bone marrow transplantation from her sister. She remains in remission for 5 months from the time of transplantation to the present. CONCLUSIONS sHLH, which may cause maternal and fetal death, should be carefully considered in critically ill pregnant women, particularly those presenting with continuous fever and cytopenia.
Collapse
Affiliation(s)
- Masaya Takahashi
- Department of Obstetrics and Gynecology, Juntendo University Urayasu Hospital, Chiba, Urayasu City, Japan
| | - Shintaro Makino
- Department of Obstetrics and Gynecology, Juntendo University Urayasu Hospital, Chiba, Urayasu City, Japan.
| | - Hiroko Iizuka
- Department of Hematology, Juntendo University Urayasu Hospital, Urayasu City, Chiba, Japan
| | - Masaaki Noguchi
- Department of Hematology, Juntendo University Urayasu Hospital, Urayasu City, Chiba, Japan
| | - Koyo Yoshida
- Department of Obstetrics and Gynecology, Juntendo University Urayasu Hospital, Chiba, Urayasu City, Japan
| |
Collapse
|
14
|
Marichannegowda MH, Mengual M, Kumar A, Giorgi EE, Tu JJ, Martinez DR, Romero-Severson EO, Li X, Feng L, Permar SR, Gao F. Different evolutionary pathways of HIV-1 between fetus and mother perinatal transmission pairs indicate unique immune selection in fetuses. Cell Rep Med 2021; 2:100315. [PMID: 34337555 PMCID: PMC8324465 DOI: 10.1016/j.xcrm.2021.100315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/12/2021] [Accepted: 05/18/2021] [Indexed: 11/04/2022]
Abstract
Study of evolution and selection pressure on HIV-1 in fetuses will lead to a better understanding of the role of immune responses in shaping virus evolution and vertical transmission. Detailed genetic analyses of HIV-1 env gene from 12 in utero transmission pairs show that most infections (67%) occur within 2 months of childbirth. In addition, the env sequences from long-term-infected fetuses are highly divergent and form separate phylogenetic lineages from their cognate maternal viruses. Host-selection sites unique to neonate viruses are identified in regions frequently targeted by neutralizing antibodies and T cell immune responses. Identification of unique selection sites in the env gene of fetal viruses indicates that the immune system in fetuses is capable of exerting selection pressure on viral evolution. Studying selection and evolution of HIV-1 or other viruses in fetuses can be an alternative approach to investigate adaptive immunity in fetuses.
Collapse
Affiliation(s)
| | - Michael Mengual
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Amit Kumar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Elena E. Giorgi
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, NM 87544, USA
| | - Joshua J. Tu
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - David R. Martinez
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Xiaojun Li
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sallie R. Permar
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Feng Gao
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| |
Collapse
|
15
|
Mou D, Ding D, Yan H, Qin B, Dong Y, Li Z, Che L, Fang Z, Xu S, Lin Y, Zhuo Y, Li J, Huang C, Zou Y, Li L, Briens M, Wu D, Feng B. Maternal supplementation of organic selenium during gestation improves sows and offspring antioxidant capacity and inflammatory status and promotes embryo survival. Food Funct 2021; 11:7748-7761. [PMID: 32794529 DOI: 10.1039/d0fo00832j] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Selenium (Se) is an essential trace element in humans and sows, having a biological function mediated in part by its incorporation into selenoproteins. This study was conducted to investigate the effects of maternal 2-hydroxy-4-methylselenobutanoic acid (HMSeBA), an organic Se source, on reproductive performance, antioxidant capacity and inflammatory status of sows and their offspring. Forty-three Landrace × Yorkshire sows were randomly allocated to receive one of the following three diets during gestation: control diet (control, basal diet, n = 15), sodium selenite (Na2SeO3) supplemented diet (Na2SeO3, basal diet + Na2SeO3 at 0.3 mg Se per kg, n = 13), and HMSeBA supplemented diet (HMSeBA, basal diet + HMSeBA at 0.3 mg Se per kg, n = 15). Blood samples of sows and piglets, placentas and piglet liver samples were analyzed for selenium status, antioxidant capacity and inflammatory cytokines. Results showed that, as compared to the control group, HMSeBA supplementation increased the number of born alive piglets and plasma concentrations of total selenium and selenoprotein P in both sows and piglets. Besides, the activities of antioxidant enzymes in the blood of sows, umbilical cord and piglets, placentas and piglets' liver were increased by dietary HMSeBA supplementation as compared to the control group, while malondialdehyde concentration (p < 0.05) was decreased in the blood of sows, umbilical cord and newborn piglets. In addition, maternal HMSeBA intake during gestation up-regulated antioxidant-related selenoprotein gene expression in the placenta (GPx2, GPx3, p < 0.05) and in the liver of newborn piglets (GPx1, GPx2, GPx3, TXNRD2, p < 0.05). Moreover, as compared to the control group, sows and newborn piglets in the Na2SeO3 and HMSeBA groups had a lower serum interleukin-6 (p < 0.05) concentration, and placentas in the HMSeBA group had lower IL-1β, IL-6 and IL-8 gene expression (p < 0.05). In conclusion, maternal supplementation of HMSeBA during pregnancy improved antioxidant capacities and reduced the inflammation level in mater, placenta, and fetus. This finding may highlight the important role of selenoproteins (especially GPXs) in preventing negative consequences of over-production of free radicals and inflammatory cytokines during gestation and at births.
Collapse
Affiliation(s)
- Daolin Mou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China and Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Dajiang Ding
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Hui Yan
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Binting Qin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yanpeng Dong
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zhen Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jian Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yuanfeng Zou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Lixia Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | | | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China and Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China. and Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China and Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
16
|
Rossouw E, Brauer M, Meyer P, du Plessis NM, Avenant T, Mans J. Virus Etiology, Diversity and Clinical Characteristics in South African Children Hospitalised with Gastroenteritis. Viruses 2021; 13:v13020215. [PMID: 33573340 PMCID: PMC7911269 DOI: 10.3390/v13020215] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Viral gastroenteritis remains a major cause of hospitalisation in young children. This study aimed to determine the distribution and diversity of enteric viruses in children ≤5 years, hospitalised with gastroenteritis at Kalafong Provincial Tertiary Hospital, Pretoria, South Africa, between July 2016 and December 2017. METHODS Stool specimens (n = 205) were screened for norovirus GI and GII, rotavirus, sapovirus, astrovirus and adenovirus by multiplex RT-PCR. HIV exposure and FUT2 secretor status were evaluated. Secretor status was determined by FUT2 genotyping. RESULTS At least one gastroenteritis virus was detected in 47% (96/205) of children. Rotavirus predominated (46/205), followed by norovirus (32/205), adenovirus (15/205), sapovirus (9/205) and astrovirus (3/205). Norovirus genotypes GI.3, GII.2, GII.3, GII.4, GII.7, GII.12, GII.21, and rotavirus strains G1P[8], G2P[4], G2P[6], G3P[4], G3P[8], G8P[4], G8P[6], G9P[6], G9P[8] and sapovirus genotypes GI.1, GI.2, GII.1, GII.4, GII.8 were detected; norovirus GII.4[P31] and rotavirus G3P[4] predominated. Asymptomatic norovirus infection (GI.3, GI.7, GII.4, GII.6, GII.13) was detected in 22% of 46 six-week follow up stools. HIV exposure (30%) was not associated with more frequent or severe viral gastroenteritis hospitalisations compared to unexposed children. Rotavirus preferentially infected secretor children (p = 0.143) and norovirus infected 78% secretors and 22% non-secretors. CONCLUSION Rotavirus was still the leading cause of gastroenteritis hospitalisations, but norovirus caused more severe symptoms.
Collapse
Affiliation(s)
- Esmari Rossouw
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Marieke Brauer
- Immunology Laboratory, Ampath, Pretoria 0001, South Africa;
| | - Pieter Meyer
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa or
- National Health Laboratory Service, Tshwane Academic Division, Pretoria 0001, South Africa
| | - Nicolette M. du Plessis
- Department of Paediatrics, Kalafong Provincial Tertiary Hospital, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (N.M.d.P.); (T.A.)
| | - Theunis Avenant
- Department of Paediatrics, Kalafong Provincial Tertiary Hospital, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (N.M.d.P.); (T.A.)
| | - Janet Mans
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
- Correspondence: ; Tel.: +27-12-319-2660
| |
Collapse
|
17
|
Guo J, Tian P, Xu Z, Zhang H. Introduction to Environmental Harmful Factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1300:3-19. [PMID: 33523427 DOI: 10.1007/978-981-33-4187-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In this Chapter, we systematically and comprehensively described various environmental harmful factors. They were classified into four aspects: physical factors, chemical factors, biological factors, and physiological and psychological stress factors. Their classification, modes of presence, toxicity and carcinogenicity, routes of exposure to human and toxic effects on the female reproductive health were introduced. It is expected that the exposure routes could be controlled and eliminated, and the pathogenic mechanism of environmental harmful factors should be investigated and explained to protect female reproductive health.
Collapse
Affiliation(s)
- Jiarong Guo
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health, Sichuan University, Chengdu, China
| | - Peng Tian
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health, Sichuan University, Chengdu, China
| | - Zhongyan Xu
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health, Sichuan University, Chengdu, China
| | - Huidong Zhang
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Association of In Utero HIV Exposure With Obesity and Reactive Airway Disease in HIV-Negative Adolescents and Young Adults. J Acquir Immune Defic Syndr 2020; 83:126-134. [PMID: 31738195 DOI: 10.1097/qai.0000000000002235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND HIV-negative individuals with in utero HIV exposure represent an emerging population, exceeding 18 million people worldwide. Long-term clinical outcomes among HIV-exposed uninfected (HEU) individuals into adolescence and young adulthood remain unknown. SETTING US academic health system. METHODS In this observational cohort study, we leveraged a patient data registry to identify 50 HEU adolescents and young adults. We also identified 141 HIV-unexposed controls that were matched to HEU subjects up to 3:1 on age of last encounter (±2 years), birthdate (±5 years), sex, race/ethnicity, and zip code. All subjects were born since January 1, 1990, with medical records available into adolescence and young adulthood. Primary outcomes were most recent body mass index (BMI) z-score and presence of reactive airway disease (RAD). Records were manually reviewed to extract health information. RESULTS Fifty HEU adolescents and young adults (18 ± 3 years, 54% men) and 141 matched controls (19 ± 3 years, 54% men) were compared. HEU individuals had a higher BMI z-score (1.12 ± 1.08 vs. 0.73 ± 1.09, P = 0.03) and an increased prevalence of obesity (42% vs. 22%, P = 0.009) compared with controls. HEU subjects also had a higher prevalence of RAD vs. controls (40% vs. 23%, P = 0.03). These differences persisted on adjusting for demographic, socioeconomic, maternal, and birth-related factors. Maternal prenatal CD4 T-cell count was inversely associated with BMI z-score among HEU adolescents (r = -0.47, P = 0.01). CONCLUSIONS HEU adolescents and young adults exhibited a heightened prevalence of obesity and RAD compared with HIV-unexposed controls. Additional studies are needed to optimize care for the expanding population of HEU individuals transitioning to adulthood.
Collapse
|
19
|
Maternal Perinatal HIV Infection Is Associated With Increased Infectious Morbidity in HIV-exposed Uninfected Infants. Pediatr Infect Dis J 2019; 38:500-502. [PMID: 30461574 PMCID: PMC6465126 DOI: 10.1097/inf.0000000000002253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND The aging population of females with perinatally-acquired HIV (PHIV) are having their own children. HIV-exposed uninfected infants (HEU-N) born to women living with non-perinatally-acquired HIV (NPHIV) experience higher infectious morbidity compared with HIV-unexposed infants (HUU). Little is known about the infectious morbidity risk of HIV-exposed uninfected infants (HEU-P) born to PHIV women. METHODS We evaluated prevalence of infectious cause hospitalizations (ICH) during the first year of life among HEU-P, HEU-N and HUU infants in a United States (U.S) tertiary care center. Maternal HIV status was categorized as PHIV vs. NPHIV vs. HIV-uninfected. Generalized Estimating Equation models were fit to evaluate the association between maternal HIV status and infant ICH. RESULTS ICH was evaluated among 205 infants, 28 HEU-P infants, 112 HEU-N infants, and 65 HUU infants. PHIV women were younger compared with NPHIV and HIV-uninfected women (median age 22 years vs. 29 and 23 respectively, p<0.01). Overall, 21% of HEU-P, 4% of HEU-N and 12% of HUU infants experienced at least one ICH event (p<0.01) in the first year of life. After adjusting for confounders, HEU-P infants were at increased ICH risk compared with HEU-N infants [adjusted odds ratio (aOR)=7.45, 95% Confidence Interval (CI):1.58-35.04]. In sub-group analysis of HEU infants, excluding HUU infants, this relationship persisted after adjustment for maternal CD4 and HIV RNA level (aOR=10.24, 95% CI:1.66-63.31) CONCLUSIONS:: In a small U.S. cohort, HEU-P infants experienced increased ICH risk. Differences in intrauterine environments, social factors, or access to care may be important factors to assess in future larger studies.
Collapse
|
20
|
Abstract
BACKGROUND HIV-exposed but uninfected (HEU) children may be at an increased risk of impaired growth when compared with their HIV-unexposed and uninfected (HUU) counterparts. We compared the growth patterns of HEU to HUU children in Nigeria. METHODS Pregnant women with and without HIV infection were enrolled at the Plateau State Specialist Hospital, Jos, Nigeria. Infants born to these mothers were recruited at birth and the mother-infant pairs followed up for 18 months. Weight, length and head circumference of the infants were measured at each visit. Age- and sex-standardized Z scores were generated for each anthropometric measure using the World Health Organization Child Growth Standards. Children with length-for-age, weight-for-age and weight-for-length Z scores <-2 were classified as stunted, underweight and wasted, respectively. RESULTS Of 415 children (307 HEU and 108 HUU) recruited for this study, 117 (28.4%), 9 (2.2%) and 32 (7.8%) infants were stunted, underweight and wasted, respectively, at birth. In a multivariable longitudinal analysis, the odds of stunting were higher among HEU as compared with HUU children [adjusted odds ratio: 2.4 (95% confidence interval: 1.4-4.1)]. Similarly, odds of being underweight were higher among the HEU children [adjusted odds ratio: 1.6 (95% confidence interval: 1.1-2.2)]. CONCLUSIONS Linear and ponderal growth were more impaired among HEU as compared with HUU children in Nigeria during the first 18 months of life. Further studies are needed to explore the causal basis for these differences.
Collapse
|
21
|
Yockey LJ, Iwasaki A. Interferons and Proinflammatory Cytokines in Pregnancy and Fetal Development. Immunity 2018; 49:397-412. [PMID: 30231982 PMCID: PMC6152841 DOI: 10.1016/j.immuni.2018.07.017] [Citation(s) in RCA: 310] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/13/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022]
Abstract
Successful pregnancy requires carefully-coordinated communications between the mother and fetus. Immune cells and cytokine signaling pathways participate as mediators of these communications to promote healthy pregnancy. At the same time, certain infections or inflammatory conditions in pregnant mothers cause severe disease and have detrimental impacts on the developing fetus. In this review, we examine evidence for the role of maternal and fetal immune responses affecting pregnancy and fetal development, both under homeostasis and following infection. We discuss immune responses that are necessary to promote healthy pregnancy and those that lead to congenital disorders and pregnancy complications, with a particular emphasis on the role of interferons and cytokines. Understanding the contributions of the immune system in pregnancy and fetal development provides important insights into the pathogenesis underlying maternal and fetal diseases and sheds insights on possible targets for therapy.
Collapse
Affiliation(s)
- Laura J Yockey
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
22
|
Wilcox CR, Jones CE. Beyond Passive Immunity: Is There Priming of the Fetal Immune System Following Vaccination in Pregnancy and What Are the Potential Clinical Implications? Front Immunol 2018; 9:1548. [PMID: 30061881 PMCID: PMC6054988 DOI: 10.3389/fimmu.2018.01548] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023] Open
Abstract
Infection is responsible for over half a million neonatal deaths worldwide every year, and vaccination in pregnancy is becoming increasingly recognized as an important strategy for the protection of young infants. Increasing evidence suggests that exposure to maternal infection in utero may "prime" the developing immune system, even in the absence of infant infection. It is also possible that in utero priming may occur following maternal vaccination, with antigen-specific cellular immune responses detectable in utero and at birth. However, this remains a topic of some controversy. This review focuses on the evidence for in utero priming and the clinical implications for vaccination in pregnancy, considering whether in utero priming following vaccination could provide protection independent of antibody-mediated passive immunity, the possible effects of vaccination on subsequent infant vaccinations, their potential "non-specific" effects, and how the design and timing of vaccination might affect prenatal priming. Looking forward, we describe other possible options for quantifying antigen-specific cellular responses, including MHC tetramers, novel proliferation and cytokine-based assays, and animal models. Together, these may help us address future research questions and establish more robust evidence of fetal immune system priming.
Collapse
Affiliation(s)
- Christopher R. Wilcox
- NIHR Clinical Research Facility, Southampton Centre for Biomedical Research, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Christine E. Jones
- Faculty of Medicine, Institute for Life Sciences, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| |
Collapse
|
23
|
Sampaio AS, Vasconcelos ALRD, Morais CNLD, Diniz GTN, Figueiredo ALDC, Montenegro SML. Social conditions and immune response in human immunodeficiency virus-seropositive pregnant women: a cross-sectional study in Brazil. Rev Soc Bras Med Trop 2018. [PMID: 29513838 DOI: 10.1590/0037-8682-0107-2017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION The functioning of the immune system during pregnancy is altered in both human immunodeficiency virus (HIV)-infected and uninfected women. Unfavorable socioeconomic conditions have been indicative of higher morbidity and mortality and worsening of the immune system. The aim of this study was to correlate social status with levels of interleukin (IL)-10 (non-inflammatory) and interferon-gamma (IFN-γ; inflammatory) cytokines. METHODS A cross-sectional study was conducted with three groups of women: 33 pregnant HIV-infected (G1); 40 non-pregnant, HIV-infected (G2); and 35 pregnant, HIV-uninfected. To measure the social status, a compound indicator called the social status index (SSI), was established using sociodemographic variables (i.e., education level, housing conditions, per capita income, and habitation and sanitary conditions). RESULTS The HIV-infected women had a higher proportion of unfavorable SSI (73% and 75% of G1 and G2, respectively). There were significantly lower IL-10 levels in the G1 group with both unfavorable and favorable SSI than in the other groups. No significant difference in IFN-γ levels was observed among groups. However, the G1 group had higher IFN-γ values among both favorable and unfavorable SSI groups. CONCLUSIONS Higher rates of unfavorable conditions, including lower education levels, IL-10 levels, and a trend for higher IFN-γ levels, were identified among HIV-infected women, pregnant and non-pregnant. These factors may interfere in health care and lead to poor outcomes during pregnancy. Therefore, we suggest that health policies could be created to specifically address these factors in this population.
Collapse
Affiliation(s)
- Aletheia Soares Sampaio
- Departamento de Saúde Coletiva, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, PE, Brasil.,Serviço de Atendimento Especializado em HIV/AIDS, Centro Integrado de Saúde Amaury de Medeiros, Universidade de Pernambuco, Recife, PE, Brasil
| | | | | | - George Tadeu Nunes Diniz
- Departamento de Saúde Coletiva, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, PE, Brasil
| | - Anna Lígia de Castro Figueiredo
- Serviço de Atendimento Especializado em HIV/AIDS, Centro Integrado de Saúde Amaury de Medeiros, Universidade de Pernambuco, Recife, PE, Brasil
| | - Sílvia Maria Lucena Montenegro
- Departamento de Imunologia e Imunopatologia Celular, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, PE, Brasil
| |
Collapse
|
24
|
Maloupazoa Siawaya AC, Mvoundza Ndjindji O, Kuissi Kamgaing E, Mveang-Nzoghe A, Mbani Mpega CN, Leboueny M, Kengue Boussougou R, Mintsa Ndong A, Essone PN, Djoba Siawaya JF. Altered Toll-Like Receptor-4 Response to Lipopolysaccharides in Infants Exposed to HIV-1 and Its Preventive Therapy. Front Immunol 2018; 9:222. [PMID: 29491865 PMCID: PMC5817973 DOI: 10.3389/fimmu.2018.00222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/26/2018] [Indexed: 11/13/2022] Open
Abstract
Pathogen sensing and recognition through pattern recognition receptors, and subsequent production of pro-inflammatory cytokines, is the cornerstone of the innate immune system. Despite the fact that HIV-exposed uninfected (HEU) infants are prone to serious bacterial infections, no study has focused on the functionality of their bacteria recognition system. This is the first study to investigate baseline levels of three critically important immune response molecules in this population: complement component (C)-3, toll-like receptor (TLR)-4, and C-reactive protein (CRP). We enrolled 16 HEU and 6 HIV-unexposed (HU) infants. TLR4 function was investigated by stimulating whole blood with increasing concentrations of TLR4-agonist ultrapure lipopolysaccharides. TLR4/TLR4-agonist dose response were assessed by measuring IL-6 secretion. Complement C3 and CRP were measured by photo spectrometry. Data showed no significant differences in baseline concentration of CRP between HEU and HU infants. Complement C3 was significantly higher in HEU infants than HU infants. TLR4 anergy was observed in 7 of 12 HEU infants, whereas the rest of HEU infants (n = 4) and the control HU infants tested (n = 3) showed responsive TLR4. None of the HEU infants investigated in this study had severe infections in the year after their birth. In conclusion, TLR4 anergy can occur in HEU infants without necessarily translating to increased vulnerability to infectious diseases.
Collapse
Affiliation(s)
- Anicet Christel Maloupazoa Siawaya
- Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori (CHUMEFJE), Libreville, Gabon.,Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique à Libreville (LNSP), Libreville, Gabon
| | - Ofilia Mvoundza Ndjindji
- Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori (CHUMEFJE), Libreville, Gabon.,Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique à Libreville (LNSP), Libreville, Gabon
| | - Eliane Kuissi Kamgaing
- Département de Pédiatrie, Université des Sciences de la Santé d'Owendo (USS), Owendo, Gabon.,Service de Néonatologie, Centre Hospitalier Universitaire de Libreville (CHUL), Libreville, Gabon
| | - Amandine Mveang-Nzoghe
- Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori (CHUMEFJE), Libreville, Gabon.,Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique à Libreville (LNSP), Libreville, Gabon
| | - Chérone Nancy Mbani Mpega
- Département de Chimie, Faculté des Sciences, Université des sciences et techniques de Masuku, Franceville, Gabon
| | - Marielle Leboueny
- Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori (CHUMEFJE), Libreville, Gabon.,Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique à Libreville (LNSP), Libreville, Gabon
| | | | - Armel Mintsa Ndong
- Unité de Virologie, Laboratoire National de Santé Publique à Libreville (LNSP), Libreville, Gabon
| | - Paulin N Essone
- Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique à Libreville (LNSP), Libreville, Gabon.,Centre de Recherche Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Joel Fleury Djoba Siawaya
- Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori (CHUMEFJE), Libreville, Gabon.,Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique à Libreville (LNSP), Libreville, Gabon
| |
Collapse
|
25
|
Jennewein MF, Abu-Raya B, Jiang Y, Alter G, Marchant A. Transfer of maternal immunity and programming of the newborn immune system. Semin Immunopathol 2017; 39:605-613. [PMID: 28971246 DOI: 10.1007/s00281-017-0653-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022]
Abstract
As placental mammals, the pregnant women and the fetus have intense and prolonged interactions during gestation. There is increasing evidence that multiple molecular as well as cellular components originating in pregnant women are transferred to the fetus. The transfer of maternal antibodies has long been recognized as a central component of newborn immunity against pathogens. More recent studies indicate that inflammatory mediators, micronutrients, microbial products and maternal cells are transferred in utero and influence the fetal immune system. Together, these multiple signals are likely to form a complex network of interactions that program the neonatal immune system and tune its homeostatic regulation. Maternal disorders, in particular infectious diseases, modify these signals and may thereby alter immunity in early life. Understanding maternal programming of the newborn immune system could provide a basis for interventions promoting child health.
Collapse
Affiliation(s)
| | - Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital, Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Yiwei Jiang
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Charleroi, Belgium
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Charleroi, Belgium.
| |
Collapse
|
26
|
Schoeman JC, Moutloatse GP, Harms AC, Vreeken RJ, Scherpbier HJ, Van Leeuwen L, Kuijpers TW, Reinecke CJ, Berger R, Hankemeier T, Bunders MJ. Fetal Metabolic Stress Disrupts Immune Homeostasis and Induces Proinflammatory Responses in Human Immunodeficiency Virus Type 1- and Combination Antiretroviral Therapy-Exposed Infants. J Infect Dis 2017. [PMID: 28633455 PMCID: PMC5853663 DOI: 10.1093/infdis/jix291] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Increased morbidity and fetal growth restriction are reported in uninfected children born to human immunodeficiency virus type 1 (HIV-1)-infected women treated with antiretroviral (ARV) therapy. Viruses and/or pharmacological interventions such as ARVs can induce metabolic stress, skewing the cell's immune response and restricting (cell) growth. Novel metabolomic techniques provided the opportunity to investigate the impact of fetal HIV-1 and combination ARV therapy (cART) exposure on the infants' immune metabolome. Peroxidized lipids, generated by reactive oxygen species, were increased in cART/HIV-1-exposed infants, indicating altered mitochondrial functioning. The lipid metabolism was further dysregulated with increased triglyceride species and a subsequent decrease in phospholipids in cART/HIV-1-exposed infants compared to control infants. Proinflammatory immune mediators, lysophospholipids as well as cytokines such as CXCL10 and CCL3, were increased whereas anti-inflammatory metabolites from the cytochrome P450 pathway were reduced in cART/HIV-1-exposed infants. Taken together, these data demonstrate that the fetal metabolism is impacted by maternal factors (cART and HIV-1) and skews physiological immune responses toward inflammation in the newborn infant.
Collapse
Affiliation(s)
- Johannes C Schoeman
- Department of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Gontse P Moutloatse
- Centre for Human Metabolomics, Faculty of Natural Sciences, North-West University, Potchefstroom, South Africa
| | - Amy C Harms
- Department of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Rob J Vreeken
- Department of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Henriette J Scherpbier
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital
| | | | - Taco W Kuijpers
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital
| | - Carools J Reinecke
- Centre for Human Metabolomics, Faculty of Natural Sciences, North-West University, Potchefstroom, South Africa
| | - Ruud Berger
- Department of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Thomas Hankemeier
- Department of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Madeleine J Bunders
- Department of Experimental Immunology.,Emma Children's Hospital, Academic Medical Center, University of Amsterdam, The Netherlands.,Research Unit Virus Immunology, Heinrich-Pette-Institute, Hamburg, Germany
| |
Collapse
|
27
|
Strickland N, Müller TL, Berkowitz N, Goliath R, Carrington MN, Wilkinson RJ, Burgers WA, Riou C. Characterization of Mycobacterium tuberculosis-Specific Cells Using MHC Class II Tetramers Reveals Phenotypic Differences Related to HIV Infection and Tuberculosis Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:ji1700849. [PMID: 28794233 PMCID: PMC5624500 DOI: 10.4049/jimmunol.1700849] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/20/2017] [Indexed: 12/21/2022]
Abstract
A major challenge for the development of an effective vaccine against tuberculosis (TB) is that the attributes of protective CD4+ T cell responses are still elusive for human TB. Infection with HIV type 1 is a major risk factor for TB, and a better understanding of HIV-induced alterations of Mycobacterium tuberculosis-specific CD4+ T cells that leads to failed host resistance may provide insight into protective T cell immunity to TB. A total of 86 participants from a TB-endemic setting, either HIV-infected or uninfected and with latent or active TB (aTB), were screened using M.tuberculosis-specific MHC class II tetramers. We examined the phenotype as well as function of ex vivo M. tuberculosis-specific tetramer+CD4+ T cells using flow cytometry. The numbers of M. tuberculosis-specific tetramer+CD4+ T cells were relatively well maintained in HIV-infected persons with aTB, despite severe immunodeficiency. However, although HIV-uninfected persons with latent TB infection exhibited ex vivo M. tuberculosis-specific CD4+ T cells predominantly of a CXCR3+CCR6+CCR4- (Th1*) phenotype, aTB or HIV infection was associated with a contraction of this subset. Nevertheless, in individuals with aTB and/or HIV infection, circulating ex vivo M. tuberculosis-specific CD4+ T cells did not display defects in exhaustion or polyfunctionality compared with healthy HIV-uninfected individuals with latent TB infection. Collectively, these data suggest that increased susceptibility to TB disease could be related to a loss of circulating Th1* CD4+ T cells rather than major changes in the number or function of circulating CD4+ T cells.
Collapse
Affiliation(s)
- Natalie Strickland
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa
| | - Tracey L Müller
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa
| | - Natacha Berkowitz
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925 Cape Town, South Africa
- Wellcome Center for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925 Cape Town, South Africa
| | - Rene Goliath
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925 Cape Town, South Africa
- Wellcome Center for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925 Cape Town, South Africa
| | - Mary N Carrington
- Cancer and Inflammation Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
- Ragon Institute of MGH, MIT and Harvard, Boston, MA 02139
| | - Robert J Wilkinson
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925 Cape Town, South Africa
- Wellcome Center for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925 Cape Town, South Africa
- Department of Medicine, Imperial College London, W2 1PG London, United Kingdom; and
- The Francis Crick Institute, NW1 2AT London, United Kingdom
| | - Wendy A Burgers
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925 Cape Town, South Africa
| | - Catherine Riou
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, 7925 Cape Town, South Africa;
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925 Cape Town, South Africa
| |
Collapse
|
28
|
Novel genetic associations and gene-gene interactions of chemokine receptor and chemokine genetic polymorphisms in HIV/AIDS. AIDS 2017; 31:1235-1243. [PMID: 28358741 DOI: 10.1097/qad.0000000000001491] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate the influence of candidate polymorphisms on chemokine receptor/ligand genes on HIV infection and AIDS progression (HIV/AIDS). DESIGN Fifteen polymorphisms of the CCR3, CCR4, CCR5, CCR6, CCR8, CXCR3, CXCR6, CCL20, CCL22 and CXCL10 genes were analysed in 206 HIV-positive patients classified as rapid progressors (n = 40), or nonrapid progressors (n = 166), and in 294 HIV-seronegative patients. METHODS The polymorphisms were genotyped using minisequencing. Genetic models were tested using binomial logistic regression; nonparametric multifactor dimensionality reduction (MDR) was used to detect gene-gene interactions. RESULTS The CCR3 rs3091250 [TT, adjusted odds ratio (AOR): 2.147, 95% confidence interval (CI) 1.076-4.287, P = 0.030], CCR8 rs2853699 (GC/CC, AOR: 1.577, 95% CI 1.049-2.371, P = 0.029), CXCL10 rs56061981 (CT/TT, AOR: 1.819, 95% CI 1.074-3.081, P = 0.026) and CCL22 rs4359426 (CA/AA, AOR: 1.887, 95% CI 1.021-3.487, P = 0.043) polymorphisms were associated with susceptibility to HIV infection. The CCL20 rs13034664 (CC, OR: 0.214, 95% CI 0.063-0.730, P = 0.014) and CCL22 rs4359426 (CA/AA, OR: 2.685, 95% CI 1.128-6.392, P = 0.026) variants were associated with rapid progression to AIDS. In MDR analyses revealed that the CXCL10 rs56061981 and CCL22 rs4359426 combination was the best model, with 57% accuracy (P = 0.008) for predicting susceptibility to HIV infection. CONCLUSION Our results provide new insights into the influence of candidate chemokine receptor/ligand polymorphisms and significant evidence for gene-gene interactions on HIV/AIDS susceptibility.
Collapse
|
29
|
Evans C, Chasekwa B, Rukobo S, Govha M, Mutasa K, Ntozini R, Humphrey JH, Prendergast AJ. Cytomegalovirus Acquisition and Inflammation in Human Immunodeficiency Virus-Exposed Uninfected Zimbabwean Infants. J Infect Dis 2017; 215:698-702. [PMID: 28011912 PMCID: PMC5388301 DOI: 10.1093/infdis/jiw630] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022] Open
Abstract
Cytomegalovirus (CMV) acquisition and inflammation were evaluated in 231 human immunodeficiency virus (HIV)–exposed uninfected (HEU) and 100 HIV-unexposed Zimbabwean infants aged 6 weeks. The HEU and HIV-unexposed infants had a similarly high prevalence of CMV (81.4% vs 74.0%, respectively; P = .14), but HEU infants had higher CMV loads (P = .005) and >2-fold higher C-reactive protein (CRP) concentrations (P < .0001). The CMV-positive HEU infants had higher CRP than the CMV-negative HEU infants; this association disappeared after adjusting for maternal HIV load. Overall, CMV acquisition is high in early life, but HEU infants have higher CMV loads and a proinflammatory milieu, which may be driven partly by maternal HIV viremia.
Collapse
Affiliation(s)
- Ceri Evans
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe.,Blizard Institute, Queen Mary University of London, London, UK
| | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Sandra Rukobo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Margaret Govha
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe.,Blizard Institute, Queen Mary University of London, London, UK.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
30
|
McFarland EJ, Powell TM, Onyango-Makumbi C, Zhang W, Melander K, Naluyima P, Okurut S, Eller MA, Fowler MG, Janoff EN. Ontogeny of CD4+ T Lymphocytes With Phenotypic Susceptibility to HIV-1 During Exclusive and Nonexclusive Breastfeeding in HIV-1-Exposed Ugandan Infants. J Infect Dis 2017; 215:368-377. [PMID: 27932619 PMCID: PMC5722036 DOI: 10.1093/infdis/jiw553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/23/2016] [Indexed: 01/17/2023] Open
Abstract
Background Among infants exposed to human immunodeficiency virus (HIV) type 1, mixed breastfeeding is associated with higher postnatal HIV-1 transmission than exclusive breastfeeding, but the mechanisms of this differential risk are uncertain. Methods HIV-1-exposed Ugandan infants were prospectively assessed during the first year of life for feeding practices and T-cell maturation, intestinal homing (β7hi), activation, and HIV-1 coreceptor (CCR5) expression in peripheral blood. Infants receiving only breast milk and those with introduction of other foods before 6 months were categorized as exclusive and nonexclusive, respectively. Results Among CD4+ and CD8+ T cells, the expression of memory, activation, and CCR5 markers increased rapidly from birth to week 2, peaking at week 6, whereas cells expressing the intestinal homing marker increased steadily in the central memory (CM) and effector memory T cells over 48 weeks. At 24 weeks, when feeding practices had diverged, nonexclusively breastfed infants showed increased frequencies and absolute counts of β7hi CM CD4+ and CD8+ T cells, including the HIV-1-targeted cells with CD4+β7hi/CCR5+ coexpression, as well as increased activation. Conclusions The T-cell phenotype associated with susceptibility to HIV-1 infection (CCR5+, gut-homing, CM CD4+ T cells) was preferentially expressed in nonexclusively breastfed infants, a group of infants at increased risk for HIV-1 acquisition.
Collapse
Affiliation(s)
- Elizabeth J McFarland
- Departments of Pediatrics
- Mucosal and Vaccine Research Program Colorado, Infectious Diseases. University of Colorado–Anschutz Medical Campus, Aurora
| | | | | | - Weiming Zhang
- Department of Biostatistics and Informatics, Colorado School of Public Health
| | | | | | - Samuel Okurut
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Michael A Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Springs
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda,
| | - Mary Glenn Fowler
- Johns Hopkins University School of Medicine, Baltimore, Maryland
- Makerere University–Johns Hopkins University Research Collaboration
| | - Edward N Janoff
- Medicine
- Mucosal and Vaccine Research Program Colorado, Infectious Diseases. University of Colorado–Anschutz Medical Campus, Aurora
- Denver Veterans Affairs Medical Center, Colorado
| |
Collapse
|
31
|
A Prospective Cohort Study of Common Childhood Infections in South African HIV-exposed Uninfected and HIV-unexposed Infants. Pediatr Infect Dis J 2017; 36:e38-e44. [PMID: 28081048 PMCID: PMC5242219 DOI: 10.1097/inf.0000000000001391] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Much evidence of HIV-exposed uninfected (HEU) infant infectious morbidity predates availability of maternal combination antiretroviral therapy and does not control for universal risk factors (preterm birth, low birth weight, suboptimal breastfeeding and poverty). METHODS This prospective cohort study identified HIV-infected and HIV-uninfected mothers and their newborns from South African community midwife unit. The primary outcome, infectious cause hospitalization or death before 6 months of age, was compared between HEU and HIV-unexposed (HU) infants and classified for type and severity using validated study-specific case definitions. Adjusted odds ratios (aORs) were calculated by logistic regression including stratified analyses conditioned on breastfeeding. RESULTS One hundred and seventy-six (94 HEU and 82 HU) mother-infant pairs were analyzed. HIV-infected mothers were older (median, 27.8 vs. 24.7 years; P < 0.01) and HU infants more often breastfed (81/82 vs. 35/94; P < 0.001). Groups were similar for maternal education, antenatal course, household characteristics, birth weight, gestational age and immunizations. The primary outcome occurred in 17 (18%) HEU and 10 (12%) HU infants [aOR, 1.45; 95% confidence interval (CI): 0.44-4.55]. In stratified analysis restricted to breastfed infants, the aOR for hospitalization due to very severe infection or death was 4.2 (95% CI: 1.00-19.2; P = 0.05) for HEU infants. Hospitalization for diarrhea was more common in HEU than HU infants [8/94 (8.5%) vs. 1/82 (1.2%); P = 0.04]. CONCLUSION The difference between HEU and HU infants in the probability of infectious cause hospitalization or death in the first 6 months of life was not significant. However, among breastfed infants, severe infectious morbidity occurred more often in HEU than HU infants.
Collapse
|
32
|
Slogrove AL, Frigati L, Gray DM. Maternal HIV and Paediatric Lung Health. Paediatr Respir Rev 2017; 21:47-53. [PMID: 27665511 DOI: 10.1016/j.prrv.2016.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/11/2016] [Indexed: 10/21/2022]
Abstract
With improved prevention of mother to child transmission of HIV, paediatric HIV disease is less common. However, the number of HIV exposed but uninfected infants is growing. Exposure to maternal HIV impacts infant respiratory health through an increase in known risk factors such as increased preterm birth and low birth weight, suboptimal breastfeeding, increased psychosocial stressors and increased exposure to infective pathogens. Exposure to the HIV virus and altered maternal immune environment result in immunologic changes in the infant that may contribute to respiratory disease risk. HIV exposed infants are at increased risk for severe pneumonia with poorer outcomes compared to unexposed infants. Maternal ART and optimal nutrition, including breastfeeding in high infective disease burden settings, reduce morbidity and mortality in HIV exposed infants and should be prioritized. The impact of exposure to maternal HIV on normal lung growth and risk for chronic respiratory disease is unknown and warrants further investigation.
Collapse
Affiliation(s)
- A L Slogrove
- Center for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town, South Africa.
| | - L Frigati
- Division of Paediatric Infectious Diseases, Department of Paediatrics and Child Health, Tygerberg Children's Hospital, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa.
| | - D M Gray
- Division Pulmonology, Department of Paediatrics and Child Health, University of Cape Town, Red Cross War Memorial Children's Hospital, Klipfontein Road, Cape Town, 7700, South Africa.
| |
Collapse
|
33
|
Maternal HIV infection alters the immune balance in the mother and fetus; implications for pregnancy outcome and infant health. Curr Opin HIV AIDS 2016; 11:138-45. [PMID: 26679415 DOI: 10.1097/coh.0000000000000239] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW With the rapid roll-out of combination antiretroviral therapy to prevent mother-to-child transmission of HIV, there is an annual increase in the number of uninfected infants born to HIV-infected women. Although the introduction of combination antiretroviral therapy has vastly improved pregnancy outcome and the health of infants born to HIV-infected women, concerns remain regarding the impact the maternal HIV infection on the pregnancy outcome and the health of HIV-exposed uninfected infants. RECENT FINDINGS Maternal HIV infection is associated with negative pregnancy outcomes such as low birth weight. In addition, an increased susceptibility to infections is reported in HIV-exposed uninfected infants compared with infants born to uninfected women. Studies have shown that HIV-exposure affects the maternal/fetal unit, with increase of proinflammatory cytokine produced by placental cells, as well as altered infant immune responses. These changes could provide the underlying conditions for negative pregnancy outcomes and facilitate mother-to-child transmission of HIV in the infant. Further studies are required to understand the underlying mechanisms and investigate whether these altered infant immune responses persist and have clinical consequences beyond childhood. SUMMARY HIV infection in pregnant women is associated with altered immune responses in HIV-infected women and their offspring with clinical consequences for pregnancy outcome and the HIV-exposed uninfected infant. Further studies are required to address the origin and long-term consequences of prenatal HIV-exposure and subsequent immune activation for infant health.
Collapse
|
34
|
Abu-Raya B, Kollmann TR, Marchant A, MacGillivray DM. The Immune System of HIV-Exposed Uninfected Infants. Front Immunol 2016; 7:383. [PMID: 27733852 PMCID: PMC5039172 DOI: 10.3389/fimmu.2016.00383] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/13/2016] [Indexed: 11/16/2022] Open
Abstract
Infants born to human immunodeficiency virus (HIV) infected women are HIV-exposed but the majority remains uninfected [i.e., HIV-exposed uninfected (HEU)]. HEU infants suffer greater morbidity and mortality from infections compared to HIV-unexposed (HU) peers. The reason(s) for these worse outcomes are uncertain, but could be related to an altered immune system state. This review comprehensively summarizes the current literature investigating the adaptive and innate immune system of HEU infants. HEU infants have altered cell-mediated immunity, including impaired T-cell maturation with documented hypo- as well as hyper-responsiveness to T-cell activation. And although prevaccination vaccine-specific antibody levels are often lower in HEU than HU, most HEU infants mount adequate humoral immune response following primary vaccination with diphtheria toxoid, haemophilus influenzae type b, whole cell pertussis, measles, hepatitis B, tetanus toxoid, and pneumococcal conjugate vaccines. However, HEU infants are often found to have lower absolute neutrophil counts as compared to HU infants. On the other hand, an increase of innate immune cytokine production and expression of co-stimulatory markers has been noted in HEU infants, but this increase appears to be restricted to the first few weeks of life. The immune system of HEU children beyond infancy remains largely unexplored.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia , Vancouver, BC , Canada
| | - Tobias R Kollmann
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia , Vancouver, BC , Canada
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles , Charleroi , Belgium
| | | |
Collapse
|
35
|
Abu-Raya B, Smolen KK, Willems F, Kollmann TR, Marchant A. Transfer of Maternal Antimicrobial Immunity to HIV-Exposed Uninfected Newborns. Front Immunol 2016; 7:338. [PMID: 27630640 PMCID: PMC5005931 DOI: 10.3389/fimmu.2016.00338] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/22/2016] [Indexed: 11/13/2022] Open
Abstract
The transfer of maternal immune factors to the newborn is critical for protection from infectious disease in early life. Maternally acquired passive immunity provides protection until the infant is beyond early life's increased susceptibility to severe infections or until active immunity is achieved following infant's primary immunization. However, as reviewed here, human immunodeficiency virus (HIV) infection alters the transfer of immune factors from HIV-infected mothers to the HIV-exposed newborns and young infants. This may relate to the immune activation in HIV-infected pregnant women, associated with the production of inflammatory cytokines at the maternofetal interface associated with inflammatory responses in the newborn. We also summarize mother-targeting interventions to improve the health of infants born to HIV-infected women, such as immunization during pregnancy and reduction of maternal inflammation. Maternal immunization offers the potential to compensate for the decreased transplacentally transferred maternal antibodies observed in HIV-exposed infants. Current data suggest reduced immunogenicity of vaccines in HIV-infected pregnant women, possibly reducing the protective impact of maternal immunization for HIV-exposed infants. Fortunately, levels of antibodies appear preserved in the breast milk of HIV-infected women, which supports the recommendation to breast-feed during antiretroviral treatment to protect HIV-exposed infants.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia , Vancouver, BC , Canada
| | - Kinga K Smolen
- Institute for Medical Immunology, Université Libre de Bruxelles , Charleroi , Belgium
| | - Fabienne Willems
- Institute for Medical Immunology, Université Libre de Bruxelles , Charleroi , Belgium
| | - Tobias R Kollmann
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia , Vancouver, BC , Canada
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles , Charleroi , Belgium
| |
Collapse
|
36
|
Altfeld M, Bunders MJ. Impact of HIV-1 infection on the feto-maternal crosstalk and consequences for pregnancy outcome and infant health. Semin Immunopathol 2016; 38:727-738. [PMID: 27392971 DOI: 10.1007/s00281-016-0578-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/16/2016] [Indexed: 12/21/2022]
Abstract
Adaptation of the maternal immune system to establish maternal/fetal equilibrium is required for a successful pregnancy. Viral infections, including HIV-1 infection, can alter this maternal/fetal equilibrium, with significant consequences for pregnancy outcome, including miscarriages, impaired fetal growth, and premature delivery. Furthermore, maternal HIV-1 infection has been shown to have a long-term impact on the developing fetal immune system also when the infant is not infected with the virus. In this review, we discuss the consequences of maternal HIV-1 infection and antiretroviral therapy on pregnancy outcome and the health of the uninfected HIV-1-exposed infant.
Collapse
Affiliation(s)
- Marcus Altfeld
- Department of Virus Immunology, Heinrich-Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Madeleine J Bunders
- Department of Virus Immunology, Heinrich-Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany. .,Department of Experimental Immunology, University of Amsterdam (UvA), Academic Medical Center (AMC), Amsterdam, The Netherlands. .,Emma Childrens Hospital,UvA, AMC, Amsterdam, The Netherlands.
| |
Collapse
|
37
|
Evans C, Humphrey JH, Ntozini R, Prendergast AJ. HIV-Exposed Uninfected Infants in Zimbabwe: Insights into Health Outcomes in the Pre-Antiretroviral Therapy Era. Front Immunol 2016; 7:190. [PMID: 27375613 PMCID: PMC4893498 DOI: 10.3389/fimmu.2016.00190] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/02/2016] [Indexed: 11/13/2022] Open
Abstract
The ZVITAMBO trial recruited 14,110 mother-infant pairs to a randomized controlled trial of vitamin A between 1997 and 2000, before the availability of antiretroviral therapy for HIV prophylaxis or treatment in Zimbabwe. The HIV status of mothers and infants was well characterized through 1-2 years of follow-up, leading to the largest cohort to date of HIV-exposed uninfected (HEU) infants (n = 3135), with a suitable comparison group of HIV-unexposed infants (n = 9510). Here, we draw on 10 years of published findings from the ZVITAMBO trial. HEU infants had increased morbidity compared to HIV-unexposed infants, with 50% more hospitalizations in the neonatal period and 30% more sick clinic visits during infancy, particularly for skin infections, lower respiratory tract infections, and oral thrush. HEU children had 3.9-fold and 2.0-fold higher mortality than HIV-unexposed children during the first and second years of life, respectively, most commonly due to acute respiratory infections, diarrhea/dysentery, malnutrition, sepsis, and meningitis. Infant morbidity and mortality were strongly related to maternal HIV disease severity, and increased morbidity remained until maternal CD4 counts were >800 cells/μL. HEU infants were more likely to be premature and small-for-gestational age than HIV-unexposed infants, and had more postnatal growth failure. Here, we propose a conceptual framework to explain the increased risk of infectious morbidity, mortality, and growth failure among HEU infants, hypothesizing that immune activation and inflammation are key drivers of both infection susceptibility and growth failure. Future studies should further dissect the causes of infection susceptibility and growth failure and determine the impact of ART and cotrimoxazole on outcomes of this vulnerable group of infants in the current era.
Collapse
Affiliation(s)
- Ceri Evans
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Blizard Institute, Queen Mary University of London, London, UK
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research , Harare , Zimbabwe
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Blizard Institute, Queen Mary University of London, London, UK; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
38
|
Slogrove AL, Goetghebuer T, Cotton MF, Singer J, Bettinger JA. Pattern of Infectious Morbidity in HIV-Exposed Uninfected Infants and Children. Front Immunol 2016; 7:164. [PMID: 27199989 PMCID: PMC4858536 DOI: 10.3389/fimmu.2016.00164] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/18/2016] [Indexed: 11/13/2022] Open
Abstract
Background Almost 30% of children in Southern Africa are HIV exposed but uninfected (HEU) and experience exposures that could increase vulnerability to infectious diseases compared to HIV unexposed (HU) children. The mechanisms of HEU infant vulnerability remain ill-defined. This review seeks to appraise the existing clinical evidence of the pattern of HEU infant infectious morbidity to aid understanding of the potential mechanism of susceptibility. Methods A systematic search was conducted of scientific literature databases and conference proceedings up to December 2015 for studies comparing adequately defined HEU (in whom HIV-infection had been excluded through age-appropriate testing) and HU infants for all-cause mortality, all-cause hospitalization, or an infection-related morbidity. The systematic review was complemented by a narrative review of additional studies detailing the pattern of infectious morbidity experienced by HEU children without comparison to HU children or without conclusive exclusion of HIV-infection in HIV-exposed infants. Results Only 3 of 22 eligible identified studies were designed to primarily compare HEU and HU infants for infectious morbidity. Fourteen were conducted prior to 2009 in the context of limited antiretroviral interventions. Three patterns emerge: (1) causes of morbidity and mortality in HEU infants are consistent with the common causes of childhood morbidity and mortality (pneumonia, diarrheal disease, and bacterial sepsis) but occur with greater severity in HEU infants resulting in higher mortality, more frequent hospitalization, and more severe manifestations of disease; (2) the greatest relative difference between HEU and HU infants in morbidity and mortality occurs beyond the neonatal period, during mid-infancy, having waned by the second year of life; and (3) HEU infants are at greater risk than HU infants for invasive streptococcal infections specifically Group B Streptococcus and Streptococcus pneumonia. Conclusion To definitively understand HEU infant infectious morbidity risk, substantially larger prospective studies with appropriate HU infant comparison groups are necessary. HEU children would benefit from collaboration among researchers to achieve the quality of evidence required to improve HEU infant outcomes globally. HEU infant health and well-being, beyond avoiding HIV-infection, deserves a more prominent position in the local and international HIV research agendas.
Collapse
Affiliation(s)
- Amy L Slogrove
- Division of Paediatric Infectious Diseases, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa; School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Tessa Goetghebuer
- Department of Paediatrics, St Pierre University Hospital, Brussels, Belgium; Université Libre de Bruxelles, Brussels, Belgium
| | - Mark F Cotton
- Division of Paediatric Infectious Diseases, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University , Tygerberg , South Africa
| | - Joel Singer
- School of Population and Public Health, University of British Columbia , Vancouver, BC , Canada
| | - Julie A Bettinger
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vaccine Evaluation Center, BC Children's Hospital, University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
39
|
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
|
40
|
Abstract
PURPOSE OF REVIEW The paediatric HIV epidemic is changing. Over the past decade, new infections have substantially reduced, whereas access to antiretroviral therapy (ART) has increased. Overall this success means that numbers of children living with HIV are climbing. In addition, the problems observed in adult infection resulting from chronic inflammation triggered by persistent immune activation even following ART mediated suppression of viral replication are magnified in children infected from birth. RECENT FINDINGS Features of immune ontogeny favour low immune activation in early life, whereas specific aspects of paediatric HIV infection tend to increase it. A subset of ART-naïve nonprogressing children exists in whom normal CD4 cell counts are maintained in the setting of persistent high viremia and yet in the context of low immune activation. This sooty mangabey-like phenotype contrasts with nonprogressing adult infection which is characterized by the expression of protective HLA class I molecules and low viral load. The particular factors contributing to raised or lowered immune activation in paediatric infection, which ultimately influence disease outcome, are discussed. SUMMARY Novel strategies to circumvent the unwanted long-term consequences of HIV infection may be possible in children in whom natural immune ontogeny in early life militates against immune activation. Defining the mechanisms underlying low immune activation in natural HIV infection would have applications beyond paediatric HIV.
Collapse
Affiliation(s)
- Julia M Roider
- aDepartment of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, UK bHIV Pathogenesis Programme, The Doris Duke Medical Research Institute cKwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | | |
Collapse
|