1
|
Zhou H, Edelman B, Skolnick J. A mode of action protein based approach that characterizes the relationships among most major diseases. Sci Rep 2025; 15:9668. [PMID: 40113859 PMCID: PMC11926353 DOI: 10.1038/s41598-025-93377-8] [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: 06/03/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
Disease classification is important for understanding disease commonalities on both the phenotypical and molecular levels. Based on predicted disease mode of action (MOA) proteins, our algorithm PICMOA (Pan-disease Classification in Mode of Action Protein Space) classifies 3526 diseases across 20 clinically classified classifications (ICD10-CM major classifications). At the top level, all diseases can be classified into "infectious" and "non-infectious" diseases. Non-infectious diseases are classified into 9 classes. To demonstrate the validity of the classifications, for common pathways predicted based on MOA proteins, 77% of the top 10 most frequent pathways have literature evidence of association to their respective disease classes/subclasses. These results indicate that PICMOA will be useful for understanding common disease mechanisms and facilitating the development of drugs for a class of diseases, rather than a single disease. The MOA proteins, molecular functions, pathways for classes, and individual diseases are available at https://sites.gatech.edu/cssb/PICMOA/ .
Collapse
Affiliation(s)
- Hongyi Zhou
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA
| | - Brice Edelman
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA.
| |
Collapse
|
2
|
Johnson MJ, Lazarus SK, Bennett AE, Tovar-Salazar A, Robertson CE, Kofonow JM, Li S, McCollister B, Nunes MC, Madhi SA, Frank DN, Weinberg A. Gut microbiota and other factors associated with increased T cell regulation in HIV-exposed uninfected infants. Front Immunol 2025; 16:1533003. [PMID: 40098966 PMCID: PMC11911520 DOI: 10.3389/fimmu.2025.1533003] [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: 11/22/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Infants exposed to HIV and uninfected (HEUs) are at higher risk of infectious morbidity than HIV-unexposed uninfected infants (HUUs). Multiple immune defects of unknown origin were observed in HEUs. We hypothesized that HEUs have more regulatory and inhibitory checkpoint-expressing T cells (Treg, Tici) than HUUs, which may dampen their immune defenses against pathogens. Method We used flow cytometry to measure 25 Treg/Tici subsets in HEUs and HUUs at birth, 6, 28, and 62 weeks of life. We used maternal and infant gut microbiome data reported in a previous study to establish correlations with the Treg/Tici. Results At birth, 3 Treg subsets, including the prototypic CD4+FOXP3+ and CD4+FOXP3+CD25+, had higher frequencies in 123 HEUs than in 117 HUUs, and 3 subsets had higher frequencies in HUUs. At 28 and 62 weeks of age, 5 Treg/Tici subsets had higher proportions in HEUs than HUUs. The frequencies of the Treg/Tici subsets that diverged between HEUs and HUUs at birth correlated with differential relative abundances of bacterial taxa in the maternal gut microbiome. The Treg/Tici subsets with significantly different frequencies at subsequent visits correlated with the concurrent composition of the infant gut microbiome. In vitro, treatment of HUU peripheral blood mononuclear cells (PBMC) with bacterial taxa most abundant in HEUs expanded Treg/Tici subsets with higher frequencies in HEUs than HUUs, recapitulating the in vivo correlations. Conversely, in vitro treatment of HEU PBMC did not increase Treg/Tici frequencies. Other factors that correlated with increased Treg/Tici frequencies were low maternal CD4+ T cells in HEUs at birth and male sex in the HUUs at 28 weeks of life. Discussion This study shows that maternal and infant gut dysbiosis are central to the increase in Treg/Tici in HEUs and may be targeted by mitigating interventions.
Collapse
Affiliation(s)
- Michael J. Johnson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sarah K. Lazarus
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ashlynn E. Bennett
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Adriana Tovar-Salazar
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Charles E. Robertson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer M. Kofonow
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Shaobing Li
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Bruce McCollister
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Marta C. Nunes
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit and Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A. Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit and Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Daniel N. Frank
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Adriana Weinberg
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
3
|
Costa PRF, Guimarães NS, Lira CRN, Leite LO, de Cássia Ribeiro da Silva R, Barreto ML, Paixão ES. Growth and body composition of adolescents and young adults with perinatal HIV infection: a systematic review and meta-analysis. BMC Public Health 2025; 25:717. [PMID: 39984871 PMCID: PMC11843769 DOI: 10.1186/s12889-025-21838-w] [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: 10/04/2023] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
INTRODUCTION Approximately 1.3 million pregnancies are exposed to HIV perinatally each year, with transmission rates ranging from 4 to 21%, despite 85% antiretroviral therapy (ART) coverage among pregnant women. While ART has significantly reduced mother-to-child transmission, recent studies indicate an increased risk of chronic conditions among perinatally HIV-infected youth. There is a critical need for further research on the growth and health of these populations to inform effective health interventions. Therefore, the aim of this study is to critically evaluate observational research assessing the growth and body composition of perinatally HIV-infected adolescents and young adults. METHODS We conducted a systematic review using the MEDLINE (by PubMed), Embase, Central (by Cochrane Library), Latin American and Caribbean Health Science Information (LILACS) and Web of Science databases. The initial search was conducted on November 24, 2022, and updated on June 3, 2024. We included cohort studies that evaluated perinatally HIV-infected adolescents (aged 10 to 19 years) and young adults (aged 20 to 24 years). A meta-analysis was performed to estimate the prevalence of stunting (HAZ < -2) and underweight (BAZ < -2). Additionally, we conducted meta-analyses for the mean values of HAZ and BAZ in the population after the follow-up period, as well as for the mean change in HAZ and BAZ post-follow-up. RESULTS Our search strategy retrieved 11,017 documents. After excluding duplicates, we analyzed 9,273 titles and abstracts. A full-text review of the remaining 87 records was conducted. Following the updated search, we identified 6 publications from 4 studies, resulting in a total of 14 studies and 16 publications. Nine studies focused exclusively on adolescents, while three evaluated both children and adolescents, and three included both adolescents and young adults. We estimated a prevalence of stunting at 26% (95% CI: 0.23-0.29) and underweight at 14% (95% CI: 0.12-0.17). In this population, the mean HAZ was -1.58 (95% CI: -1.90; -1.27), and the mean BAZ was -0.34 (95% CI: -0.61; -0.06). Additionally, we identified an increase of 0.55 (95% CI: 0.07; 1.03) in mean HAZ and 0.12 (95% CI: -0.56; 0.79) in mean BAZ after the follow-up period. CONCLUSIONS Our study highlights significant growth and body composition challenges among perinatally HIV-infected adolescents and young adults, with concerning rates of stunting and underweight. Although there was a modest increase in height-for-age, persistent challenges in achieving optimal growth remain. The slight improvement in BMI is insufficient to fully address underweight concerns. The limited number of studies and their inherent limitations restrict the ability to draw consistent conclusions regarding the effects and magnitude of exposure on anthropometric outcomes. Further research is needed to better elucidate these relationships. TRIAL REGISTRATION CRD42022372837.
Collapse
Affiliation(s)
- Priscila R F Costa
- Department of Science Nutrition, Federal University of Bahia, 32 Araujo Pinho Avenue, Salvador, 40.110-907, Brazil.
- Post-Graduation Program in Food, Nutrition and Health, Federal University of Bahia, Salvador, Brazil.
- Center for Data and Knowledge Integration for Health (CIDACS), Oswaldo Cruz Foundation, Salvador, Brazil.
| | - Nathalia S Guimarães
- Department of Nutrition, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlos R N Lira
- Post-Graduation Program in Food, Nutrition and Health, Federal University of Bahia, Salvador, Brazil
| | - Luana O Leite
- Post-Graduation Program in Food, Nutrition and Health, Federal University of Bahia, Salvador, Brazil
| | - Rita de Cássia Ribeiro da Silva
- Department of Science Nutrition, Federal University of Bahia, 32 Araujo Pinho Avenue, Salvador, 40.110-907, Brazil
- Post-Graduation Program in Food, Nutrition and Health, Federal University of Bahia, Salvador, Brazil
- Center for Data and Knowledge Integration for Health (CIDACS), Oswaldo Cruz Foundation, Salvador, Brazil
| | - Maurício L Barreto
- Department of Nutrition, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Enny S Paixão
- Department of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
4
|
Mataramvura H, Jӓger J, Jordan-Paiz A, Mazengera LR, Gumbo FZ, Bunders MJ, Duri K. Phenotypic characterization of NK cells in 5-year-old children exposed to maternal HIV and antiretroviral therapy in early-life. BMC Immunol 2024; 25:82. [PMID: 39702040 DOI: 10.1186/s12865-024-00674-4] [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: 02/27/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND HIV-exposed uninfected (HEU) children are at increased risk of morbidity during the first years of life. Although the immune responses of HEU infants in early-life are relatively well described, studies of natural killer (NK) cells in older HEU children are lacking. NK cell subsets were analysed in HEU children and compared to those in HIV unexposed uninfected (HUU) children aged ~ five years. METHODS Multi-parametric flow cytometry was used to characterize peripheral blood-derived NK cell CD56, CD16, CD57, NKG2A and KIR3DL1/KIR2DL2/L3 expression, including intracellular perforin and granzyme B. NK cell subsets were compared between HEU children exposed to prenatal antiretroviral therapy (ART) from conception [long-term (HEULT)]; those exposed to ART during pregnancy [medium-term (HEUMT)] with continued exposure throughout the breastfeeding period and HUU peers. Furthermore, clinical data of the children, including sick clinic visits and hospitalizations documented in morbidity diaries from birth to 5 years were compared between HEU and HUU groups. Frequencies of CD56bright and CD56dim NK cell were correlated with these clinical parameters. RESULTS 139 children were enrolled however, 133 comprising 43 HEULT, 38 HEUMT and 52 HUU were included in the main analyses. Total NK cell, CD56bright nor CD56dim NK cell proportions differed between HEU and HUU children. However, HEULT children had lower frequencies of CD56dim NK cells compared to HEUMT children, (p = 0.002) which maintained significance after controlling for preterm birth, p = 0.012. No differences were observed between HEULT and HUU. The expressions of NKG2A, KIR3DL1/KIR2DL2/L3 and CD57 on CD56bright and CD56dim NK cells were similar between the three groups. Furthermore, the frequencies of granzyme B and perforin double positive NK cells were similar between the HUU with HEULT and HEUMT children. CD56dim NK cell counts had a significant moderate negative correlation with recurrent respiratory infections (rho=-0.38; p = 0.010) in HUU children and negatively correlated with total sick clinic visits in HEUMT (rho=-0.40, p = 0.064). CONCLUSION The proportions of total NK cell, CD56bright and CD56dim NK cells, NK cells inhibitory and differentiation surface marker expression and cytolytic granule-positive cells were similar between HEU and HUU children. These data suggest that early-life HIV/ART exposure may not result in major changes in NK cell subsets at 5 years of age.
Collapse
Affiliation(s)
- Hope Mataramvura
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, Faculty of Medicine and Health Sciences, University of Zimbabwe, UZ-FMHS), Harare, Zimbabwe.
| | - Julia Jӓger
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Ana Jordan-Paiz
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Lovemore Ronald Mazengera
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, Faculty of Medicine and Health Sciences, University of Zimbabwe, UZ-FMHS), Harare, Zimbabwe
| | | | - Madeleine J Bunders
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
- III. Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Kerina Duri
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, Faculty of Medicine and Health Sciences, University of Zimbabwe, UZ-FMHS), Harare, Zimbabwe
| |
Collapse
|
5
|
Kefale B, Jancey J, Gebremedhin AT, Nyadanu SD, Belay DG, Pereira G, Tessema GA. Risk factors of under-five and infant mortality: An umbrella review of systematic reviews and meta-analyses. J Glob Health 2024; 14:04260. [PMID: 39611446 PMCID: PMC11605776 DOI: 10.7189/jogh.14.04260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
Background Ensuring child survival is a critical global challenge, requiring a robust and comprehensive understanding of the risk factors contributing to under-five mortality (U5M). We aimed to synthesise and summarise the current available evidence on risk factors of U5M and infant mortality worldwide to inform global child health programmes. Methods We searched six major databases (Embase, Medline, Scopus, CINAHL, Web of Science, and Global Health) and repositories of systematic reviews, as well as grey literature sources to identify systematic reviews and meta-analyses that examined the associations between risk factors of U5M and infant mortality between 1 January 1990 and 4 March 2024. The quality of reviews was assessed using A Measurement Tool to Assess Systematic Reviews, Version 2 (AMSTAR 2). The strength of evidence and direction of associations was graded. Results Of 5684 records, we included 32 reviews (including five systematic reviews without meta-analysis) which comprised 1042 primary studies. We synthesised 28 and 29 unique risk factors associated with U5M and infant mortality, respectively. Although there was no convincing evidence for the risk factors, we found probable evidence of association between exclusive breastfeeding (consistent negative association), and maternal death (consistent positive association) with U5M. There was also probable evidence for the association of short (<18 months) interpregnancy intervals (less consistent negative association), pre-pregnancy maternal obesity (consistent positive association), and maternal HIV infection (consistent positive association) with infant mortality. Conclusions While the review identified a broad range of risk factors, the overall evidence for most factors associated with under-five and infant mortality was 'limited-suggestive' or 'limited and no conclusive'. Thus, further high-quality studies are required to strengthen the evidence on these risk factors. Registration PROSPERO CRD42023455542.
Collapse
Affiliation(s)
- Bereket Kefale
- Curtin School of Population Health, Curtin University, Perth, Western Australia, Australia
- Department of Reproductive Health, School of Public Health, Wollo University, Dessie, Ethiopia
| | - Jonine Jancey
- Curtin School of Population Health, Curtin University, Perth, Western Australia, Australia
- enAble Institute, Curtin University, Perth, Western Australia, Australia
| | - Amanuel T Gebremedhin
- Curtin School of Population Health, Curtin University, Perth, Western Australia, Australia
- School of Nursing and Midwifery, Edith Cowan University, Perth, Western Australia, Australia
| | | | - Daniel G Belay
- Curtin School of Population Health, Curtin University, Perth, Western Australia, Australia
| | - Gavin Pereira
- Curtin School of Population Health, Curtin University, Perth, Western Australia, Australia
- enAble Institute, Curtin University, Perth, Western Australia, Australia
| | - Gizachew A Tessema
- Curtin School of Population Health, Curtin University, Perth, Western Australia, Australia
- enAble Institute, Curtin University, Perth, Western Australia, Australia
- School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
6
|
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
|
7
|
Simeni LAK, Ekali GL, Yengo CK, Wouambo RK, Fischer J, Bessong OMA, Fokam J, Yindom LM, Nguedia JCA. KIR2DL1 gene is a surrogate marker of protection against infection-related hospitalization among HIV-1 unexposed versus exposed uninfected infants in Cameroon. BMC Immunol 2024; 25:54. [PMID: 39090586 PMCID: PMC11293080 DOI: 10.1186/s12865-024-00645-9] [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: 02/17/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND HIV-exposed uninfected infants (HEU) appear more vulnerable to infections compared to their HIV-unexposed uninfected (HUU) peers, generally attributed to poor passive immunity acquired from the mother. This may be due to some genetic factors that could alter the immune system. We thus sought to determine the distribution of Killer Cell Immunoglobulin-Like Receptors (KIRs) genes in HEU versus HUU and study their associations with the occurrence of infection-related hospitalization. METHODS A cohort study was conducted from May 2019 to April 2020 among HEU and HUU infants, including their follow-up at weeks 6, 12, 24, and 48, in reference pediatric centers in Yaoundé-Cameroon. The infant HIV status and infections were determined. A total of 15 KIR genes were investigated using the sequence-specific primer polymerase chain reaction (PCR-SSP) method. The KIR genes that were significantly associated with HIV-1 status (HEU and HUU) were analyzed for an association with infection-related hospitalizations. This was only possible if, and to the extent that, infection-related hospitalizations varied significantly according to status. Multivariate logistic regression analyses were conducted to determine the association between KIR gene content variants and HIV status, while considering a number of potential confounding factors. Furthermore, the risk was quantified using relative risk, odds ratio, and a 95% confidence interval. The Fisher exact test was employed to compare the frequency of occurrences. A p-value of less than 0.05 was considered statistically significant. RESULTS In this cohort, a total of 66 infants participated, but only 19 acquired infections requiring hospitalizations (14.81%, 04/27 HUU and 38.46%, 15/39 HEU, p = 0.037). At week 48 (39 HEU and 27 HUU), the relative risk (RR) for infection-related hospitalizations was 2.42 (95% CI: 1.028-5.823) for HEU versus HUU with OR 3.59 (1.037-12.448). KIR2DL1 gene was significantly underrepresented in HEU versus HUU (OR = 0.183, 95%CI: 0.053-0.629; p = 0.003), and the absence of KIR2DL1 was significantly associated with infection-related hospitalization (p < 0.001; aOR = 0.063; 95%CI: 0.017-0.229). CONCLUSION Compared to HUU, the vulnerability of HEU is driven by KIR2DL1, indicating the protective role of this KIR against infection and hospitalizations.
Collapse
Affiliation(s)
- Luc-Aimé Kagoué Simeni
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Buéa, Buéa, Cameroon.
- American Association of Microbiology (ASM), ASM Cameroon, Bangangté, Cameroon.
| | | | - Clauvis Kunkeng Yengo
- Department of Biochemistry, Faculty of Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Rodrigue Kamga Wouambo
- American Association of Microbiology (ASM), ASM Cameroon, Bangangté, Cameroon
- Division of Hepatology, Leipzig University Medical Centre, Leipzig, Germany
| | - Janett Fischer
- Division of Hepatology, Leipzig University Medical Centre, Leipzig, Germany
| | | | - Joseph Fokam
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Buéa, Buéa, Cameroon
- Virology Laboratory, Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | | | | |
Collapse
|
8
|
Cherkos AS, LaCourse SM, Kinuthia J, Mecha J, Enquobahrie DA, Escudero JN, John-Stewart G. Maternal breastfeeding and education impact infant growth and development more than in-utero HIV/antiretroviral therapy exposure in context of universal antiretroviral therapy. AIDS 2024; 38:537-546. [PMID: 37967230 PMCID: PMC10922740 DOI: 10.1097/qad.0000000000003785] [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] [Indexed: 11/17/2023]
Abstract
BACKGROUND Exposure to HIV and antiretroviral therapy (ART) in utero may influence infant growth and development. Most available evidence predates adoption of universal ART (Option B+ ART regimens). In a recent cohort, we compared growth and development in HIV-exposed uninfected (HEU) to HIV-unexposed (HUU) infants. DESIGN Prospective cohort study: data from Impact of Maternal HIV on Mycobacterium Tuberculosis Infection among Peripartum Women and their Infants (MiTIPS) in Western Kenya. METHODS Women were enrolled during pregnancy. Mother-infant pairs were followed until 24 months postpartum. We used multivariable linear mixed-effects models to compare growth rates [weight-for-age z score (WAZ) and height-for-age z score (HAZ)] and multivariable linear regression to compare overall development between HEU and HUU children. RESULTS About 51.8% (184/355) of the infants were HEU, 3.9% low birthweight (<2.5 kg), and 8.5% preterm (<37 gestational weeks). During pregnancy, all mothers of HEU received ART; 67.9% started ART prepregnancy, and 87.3% received 3TC/FTC, TDF, and EFV. In longitudinal analyses, HEU children did not differ significantly from HUU in growth or development ( P > 0.05 for all). In the combined HEU/HUU cohort, higher maternal education was associated with significantly better growth and development: WAZ [ β = 0.18 (95% CI 0.01-0.34)], HAZ [ β = 0.26 (95% CI 0.04-0.48)], and development [ β = 0.24 (95% CI 0.02-0.46)]. Breastfeeding was associated with significantly better HAZ [ β =0.42 (95% CI 0.19-0.66)] and development [ β =0.31 (95% CI 0.08-0.53)]. CONCLUSION HEU children in the setting of universal maternal ART had a similar growth trajectory and development to HUU children. Breastfeeding and maternal education improved children's weight, height, and overall development irrespective of maternal HIV status.
Collapse
Affiliation(s)
- Ashenafi S Cherkos
- Department of Population and Community Health, University of North Texas Health Science Center, Fort Worth, TX
| | - Sylvia M LaCourse
- Department of Epidemiology, School of Public Health
- Department of Medicine, Division of Allergy and Infectious Diseases
- Department of Global Health, School of Public Health, University of Washington, Seattle, WA, USA
| | - John Kinuthia
- Research and Programs
- Department of Obstetrics and Gynaecology, Kenyatta National Hospital, Nairobi, Kenya
| | | | | | - Jaclyn N Escudero
- Department of Global Health, School of Public Health, University of Washington, Seattle, WA, USA
| | - Grace John-Stewart
- Department of Epidemiology, School of Public Health
- Department of Medicine, Division of Allergy and Infectious Diseases
- Department of Global Health, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
9
|
Camacho-Pacheco RT, Hernández-Pineda J, Brito-Pérez Y, Plazola-Camacho N, Coronado-Zarco IA, Arreola-Ramírez G, Bermejo-Haro MY, Najera-Hernández MA, González-Pérez G, Herrera-Salazar A, Olmos-Ortiz A, Soriano-Becerril D, Sandoval-Montes C, Figueroa-Damian R, Rodríguez-Martínez S, Mancilla-Herrera I. Disturbances in the IgG Antibody Profile in HIV-Exposed Uninfected Infants Associated with Maternal Factors. J Immunol Res 2024; 2024:8815767. [PMID: 38375063 PMCID: PMC10876311 DOI: 10.1155/2024/8815767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 02/21/2024] Open
Abstract
Over the last 20 years, the incidence of vertical HIV transmission has decreased from 25%-42% to less than 1%. Although there are no signs of infection, the health of HIV-exposed uninfected (HEU) infants is notoriously affected during the first months of life, with opportunistic infections being the most common disease. Some studies have reported effects on the vertical transfer of antibodies, but little is known about the subclass distribution of these antibodies. We proposed to evaluate the total IgG concentration and its subclasses in HIV+ mothers and HEU pairs and to determine which maternal factors condition their levels. In this study, plasma from 69 HEU newborns, their mothers, and 71 control pairs was quantified via immunoassays for each IgG isotype. Furthermore, we followed the antibody profile of HEUs throughout the first year of life. We showed that mothers present an antibody profile characterized by high concentrations of IgG1 and IgG3 but reduced IgG2, and HEU infants are born with an IgG subclass profile similar to that of their maternal pair. Interestingly, this passively transferred profile could remain influenced even during their own antibody production in HEU infants, depending on maternal conditions such as CD4+ T-cell counts and maternal antiretroviral treatment. Our findings indicate that HEU infants exhibit an altered IgG subclass profile influenced by maternal factors, potentially contributing to their increased susceptibility to infections.
Collapse
Affiliation(s)
- Rodrigo T. Camacho-Pacheco
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Jessica Hernández-Pineda
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Yesenia Brito-Pérez
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Noemi Plazola-Camacho
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | | | | | - Mextli Y. Bermejo-Haro
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - M. Angel Najera-Hernández
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Gabriela González-Pérez
- Department of Physiology and Cellular Development, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Alma Herrera-Salazar
- Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, UNAM, Cuautitlán Izcalli, Mexico
| | - Andrea Olmos-Ortiz
- Immunobiochemistry Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Diana Soriano-Becerril
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Claudia Sandoval-Montes
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ricardo Figueroa-Damian
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| | - Sandra Rodríguez-Martínez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ismael Mancilla-Herrera
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico
| |
Collapse
|
10
|
Powis KM, Lebanna L, Schenkel S, Masasa G, Kgole SW, Ngwaca M, Kgathi C, Williams PL, Slogrove AL, Shapiro RL, Lockman S, Mmalane MO, Makhema JM, Jao J, Cassidy AR. Lower academic performance among children with perinatal HIV exposure in Botswana. J Int AIDS Soc 2023; 26 Suppl 4:e26165. [PMID: 37909233 PMCID: PMC10618869 DOI: 10.1002/jia2.26165] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 08/23/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION Studies have reported a higher risk of suboptimal neurodevelopment among children who are HIV-exposed uninfected (HEU) compared to children HIV-unexposed uninfected (HUU). Actual academic performance among school-aged children by HIV exposure status has not been studied. METHODS Academic performance in Mathematics, Science, English, Setswana and overall among children enrolled in the Botswana-based FLOURISH study who were attending public primary school and ranging in age from 7.1 to 14.6 years were compared by HIV exposure status using a Cochran-Mantel-Haenszel test. Lower academic performance was defined as a grade of "C" or lower (≤60%). Unadjusted and adjusted logistic regression models were fit to assess for an association between HIV exposure and lower academic performance. RESULTS Between April 2021 and December 2022, 398 children attending public primary school enrolled in the FLOURSH study, 307 (77%) were HEU. Median age was 9.4 years (IQR 8.9-10.2). Only 17.9% of children HEU were breastfeed versus 100% of children HUU. Among children HEU, 80.3% had foetal exposure to three-drug antiretroviral treatment, 18.7% to zidovudine only and 1.0% had no antiretroviral exposure. Caregivers of children HEU were older compared to caregivers of children HUU (median 42 vs. 36 years) and more likely to have no or primary education only (15.0% vs. 1.1%). In unadjusted analyses, children HEU were more likely to have lower overall academic performance compared to their children HUU (odds ratio [OR]: 1.96 [95% confidence interval (CI): 1.16, 3.30]), and lower performance in Mathematics, Science and English. The association was attenuated after adjustment for maternal education, caregiver income, breastfeeding, low birth weight and child sex (aOR: 1.86 [95% CI: 0.78, 4.43]). CONCLUSIONS In this Botswana-based cohort, primary school academic performance was lower among children HEU compared to children HUU. Biological and socio-demographic factors, including child sex, appear to contribute to this difference. Further research is needed to identify modifiable contributors, develop screening tools to identify the risk of poor academic performance and design interventions to mitigate risk.
Collapse
Affiliation(s)
- Kathleen M. Powis
- Department of Internal Medicine and PediatricsMassachusetts General HospitalBostonMassachusettsUSA
- Department of Immunology and Infectious DiseasesHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Botswana Harvard Health PartnershipGaboroneBotswana
| | - Lesedi Lebanna
- Department of Curriculum Development and EvaluationBotswana Ministry of Basic EducationGaboroneBotswana
| | - Sara Schenkel
- Department of Internal Medicine and PediatricsMassachusetts General HospitalBostonMassachusettsUSA
| | | | | | | | | | - Paige L. Williams
- Departments of Biostatistics and EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Amy L. Slogrove
- Department of Paediatrics & Child HealthFaculty of Medicine and Health SciencesStellenbosch UniversityWorcesterSouth Africa
| | - Roger L. Shapiro
- Department of Immunology and Infectious DiseasesHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Botswana Harvard Health PartnershipGaboroneBotswana
| | - Shahin Lockman
- Department of Immunology and Infectious DiseasesHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Botswana Harvard Health PartnershipGaboroneBotswana
- Division of Infectious DiseasesBrigham and Women's HospitalBostonMassachusettsUSA
| | | | | | - Jennifer Jao
- Botswana Harvard Health PartnershipGaboroneBotswana
- Department of PediatricsNorthwestern Feinberg School of MedicineChicagoIllinoisUSA
| | - Adam R. Cassidy
- Botswana Harvard Health PartnershipGaboroneBotswana
- Departments of Psychiatry & Psychology and Pediatric & Adolescent MedicineMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
11
|
Mataramvura H, Bunders MJ, Duri K. Human immunodeficiency virus and antiretroviral therapy-mediated immune cell metabolic dysregulation in children born to HIV-infected women: potential clinical implications. Front Immunol 2023; 14:1182217. [PMID: 37350953 PMCID: PMC10282157 DOI: 10.3389/fimmu.2023.1182217] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023] Open
Abstract
Commencing lifelong antiretroviral therapy (ART) immediately following HIV diagnosis (Option B+) has dramatically improved the health of HIV-infected women and their children, with the majority being of HIV-exposed children born uninfected (HEU). This success has led to an increasing population of HIV-infected women receiving ART during pregnancy and children exposed to ART in utero. Nonetheless, a small proportion of children are still infected with HIV (HEI) each year. HEI children suffer from reduced immunocompetence and host-defence, due to CD4+ T lymphocyte depletion, but also dysregulation of other immune cells including CD8+ T lymphocytes, natural killer (NK) cells, macrophages including B lymphocytes. Furthermore, although HEU children are uninfected, altered immune responses are observed and associated with increased vulnerability to infections. The mechanisms underlying immune dysregulation in HEU children remain poorly described. Building on early studies, emerging data suggests that HIV/ART exposure early in life affects cell metabolic function of HEU children. Prenatal HIV/ART exposure has been associated with dysregulation of mitochondria, including impaired DNA polymerase activity. Furthermore, dysregulation of oxidative phosphorylation (OXPHOS) causes a decreased generation of adenosine triphosphate (ATP) and increased production of reactive oxygen species (ROS), resulting in oxidative stress. These altered metabolic processes can affect immune cell viability and immune responses. Recent studies have indicated that immune-metabolic dysregulation may contribute to HIV-associated pathogenesis and clinical observations associated with HIV and ART exposure in HEU/HEI children. Given the critical role metabolic processes in immune cell functioning, immune-metabolic dysregulation in HEU and HEI children may have implications in effective host-defence responses against pathogens, as well as efficacy of standard ART regimens and future novel HIV cure approaches in HEI children. At the same time, targeting metabolic pathways of immune cells may provide safer and novel approaches for HIV cure strategies. Here, we review the current literature investigating immune-metabolic dysregulation in paediatric HIV pathogenesis.
Collapse
Affiliation(s)
- Hope Mataramvura
- Immunology Unit, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Madeleine J. Bunders
- III. Medical Department, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Kerina Duri
- Immunology Unit, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| |
Collapse
|
12
|
Nziza N, Jung W, Mendu M, Chen T, McNamara RP, Fortune SM, Franken KLMC, Ottenhoff THM, Bryson B, Ngonzi J, Bebell LM, Alter G. Maternal HIV infection drives altered placental Mtb-specific antibody transfer. Front Microbiol 2023; 14:1171990. [PMID: 37228375 PMCID: PMC10203169 DOI: 10.3389/fmicb.2023.1171990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Placental transfer of maternal antibodies is essential for neonatal immunity over the first months of life. In the setting of maternal HIV infection, HIV-exposed uninfected (HEU) infants are at higher risk of developing severe infections, including active tuberculosis (TB). Given our emerging appreciation for the potential role of antibodies in the control of Mycobacterium tuberculosis (Mtb), the bacteria that causes TB, here we aimed to determine whether maternal HIV status altered the quality of Mtb-specific placental antibody transfer. Methods Antigen-specific antibody systems serology was performed to comprehensively characterize the Mtb-specific humoral immune response in maternal and umbilical cord blood from HIV infected and uninfected pregnant people in Uganda. Results Significant differences were noted in overall antibody profiles in HIV positive and negative maternal plasma, resulting in heterogeneous transfer of Mtb-specific antibodies. Altered antibody transfer in HIV infected dyads was associated with impaired binding to IgG Fc-receptors, which was directly linked to HIV viral loads and CD4 counts. Conclusions These results highlight the importance of maternal HIV status on antibody transfer, providing clues related to alterations in transferred maternal immunity that may render HEU infants more vulnerable to TB than their HIV-unexposed peers.
Collapse
Affiliation(s)
- Nadege Nziza
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Wonyeong Jung
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Maanasa Mendu
- Department of Molecular and Cellular Biology, Harvard University, Boston, MA, United States
| | - Tina Chen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Ryan P. McNamara
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Sarah M. Fortune
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Kees L. M. C. Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bryan Bryson
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Joseph Ngonzi
- Department of Obstetrics and Gynecology, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Lisa M. Bebell
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, United States
- Center for Global Health, Massachusetts General Hospital, Boston, MA, United States
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
13
|
du Toit LDV, Prinsloo A, Steel HC, Feucht U, Louw R, Rossouw TM. Immune and Metabolic Alterations in Children with Perinatal HIV Exposure. Viruses 2023; 15:v15020279. [PMID: 36851493 PMCID: PMC9966389 DOI: 10.3390/v15020279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
With the global rollout of mother-to-child prevention programs for women living with HIV, vertical transmission has been all but eliminated in many countries. However, the number of children who are exposed in utero to HIV and antiretroviral therapy (ART) is ever-increasing. These children who are HIV-exposed-but-uninfected (CHEU) are now well recognized as having persistent health disparities compared to children who are HIV-unexposed-and-uninfected (CHUU). Differences reported between these two groups include immune dysfunction and higher levels of inflammation, cognitive and metabolic abnormalities, as well as increased morbidity and mortality in CHEU. The reasons for these disparities remain largely unknown. The present review focuses on a proposed link between immunometabolic aberrations and clinical pathologies observed in the rapidly expanding CHEU population. By drawing attention, firstly, to the significance of the immune and metabolic alterations observed in these children, and secondly, the impact of their healthcare requirements, particularly in low- and middle-income countries, this review aims to sensitize healthcare workers and policymakers about the long-term risks of in utero exposure to HIV and ART.
Collapse
Affiliation(s)
- Louise D V du Toit
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
- UP Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria 0001, South Africa
- Maternal and Infant Health Care Strategies Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
| | - Andrea Prinsloo
- UP Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria 0001, South Africa
- Maternal and Infant Health Care Strategies Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Hematology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Helen C Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Ute Feucht
- UP Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria 0001, South Africa
- Maternal and Infant Health Care Strategies Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Pediatrics, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Theresa M Rossouw
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
- UP Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria 0001, South Africa
- Maternal and Infant Health Care Strategies Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
| |
Collapse
|
14
|
Slogrove AL, de Beer ST, Kalk E, Boulle A, Cotton M, Cupido H, Laughton B, Marlow M, Mehta U, Msolo N, Myer L, Powis KM, Schoeman E, Tomlinson M, Zunza M, Williams P, Davies MA. Survival and health of children who are HIV-exposed uninfected: study protocol for the CHERISH (Children HIV-Exposed Uninfected - Research to Inform Survival and Health) dynamic, prospective, maternal-child cohort study. BMJ Open 2023; 13:e070465. [PMID: 36593001 PMCID: PMC9809249 DOI: 10.1136/bmjopen-2022-070465] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/15/2022] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION CHERISH is designed to establish a long-term sustainable system for measurement of in utero and postnatal exposures and outcomes in children who are HIV-exposed uninfected (HEU) and HIV-unexposed to compare survival, hospitalisation, growth and neurodevelopment in the Western Cape, South Africa. METHODS AND ANALYSIS During 2022-2025, the CHERISH dynamic cohort is prospectively enrolling pregnant people with and without HIV at 24-36 weeks gestation from one urban and one rural community, following mother-child pairs, including children who are HEU (target N=1200) and HIV-unexposed (target N=600) for 3 years from the child's birth. In-person visits occur at enrolment, delivery, 12 months, 24 months and 36 months with intervening 3-monthly telephone data collection. Children and mothers without HIV are tested for HIV at all in-person visits. Data on exposures and outcomes are collected from routine standardised healthcare documentation, maternal interview, measurement (growth and neurodevelopment) at in-person visits and linkage to the Western Cape Provincial Health Data Centre (survival and hospitalisation). A priori adverse birth outcomes, advanced maternal HIV and maternal mental health are considered potential mediators of outcome disparities in children who are HEU and will be evaluated as such in multivariable models appropriate for each outcome. ETHICS AND DISSEMINATION Mothers interested in joining the study are taken through a visual informed consent document for their and their child's participation, with the option to consent to anonymised de-identified data being contributed to a public data repository. All data is captured directly into an electronic database using alphanumeric identifiers devoid of identifying information. The cohort study is approved by Human Research Ethics Committees of Stellenbosch University (N20/08/084), University of Cape Town (723/2021) and Western Cape Government (WC_2021_09_007). Findings will be shared with participants, participating communities, local and provincial stakeholders, child health clinicians, researchers and policymakers at local, national and international forums and submitted for publication in peer-reviewed journals.
Collapse
Affiliation(s)
- Amy L Slogrove
- Department of Paediatrics and Child Health, Stellenbosch University Faculty of Medicine and Health Sciences, Worcester, South Africa
- Department of Global Health, Ukwanda Centre for Rural Health, Stellenbosch University Faculty of Medicine and Health Sciences, Worcester, South Africa
| | - Shani Tamlyn de Beer
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
- Division of Population Health Sciences, University of Bristol, Bristol, UK
| | - Emma Kalk
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
| | - Andrew Boulle
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
- Health Intelligence Directorate, Western Cape Provincial Government, Cape Town, South Africa
| | - Mark Cotton
- Department of Paediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Heinrich Cupido
- Department of Paediatrics and Child Health, Stellenbosch University Faculty of Medicine and Health Sciences, Worcester, South Africa
| | - Barbara Laughton
- Department of Paediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Marguerite Marlow
- Department of Global Health, Institute for Life Course Health Research, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Ushma Mehta
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
| | - Ncumisa Msolo
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
| | - Landon Myer
- Division of Epidemiology and Biostatistics, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
| | - Kathleen M Powis
- Departments of Internal Medicine and Paediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Elisma Schoeman
- Department of Paediatrics and Child Health, Stellenbosch University Faculty of Medicine and Health Sciences, Worcester, South Africa
| | - Mark Tomlinson
- Department of Global Health, Institute for Life Course Health Research, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Moleen Zunza
- Department of Global Health, Division of Epidemiology and Biostatistics, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Paige Williams
- Department of Biostatistics, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
- Health Intelligence Directorate, Western Cape Provincial Government, Cape Town, South Africa
| |
Collapse
|
15
|
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
|
16
|
Emmanuel PJ, Mansfield J, Siberry GK. Human Immunodeficiency Virus Infection: An Update for Pediatricians. Pediatr Rev 2022; 43:335-346. [PMID: 35641447 DOI: 10.1542/pir.2020-001644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Patricia J Emmanuel
- Department of Pediatrics, University of South Florida Morsani College of Medicine, Tampa, FL
| | | | - George K Siberry
- Division of Prevention Care and Treatment, Office of HIV/AIDS, US Agency for International Development, Washington, DC
| |
Collapse
|
17
|
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: 2] [Impact Index Per Article: 0.7] [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
|
18
|
Smith C, Huo Y, Patel K, Fetters K, Hegemann S, Burchett S, Van Dyke R, Weinberg A. Immunologic and Virologic Factors Associated With Hospitalization in Human Immunodeficiency Virus-Exposed, Uninfected Infants in the United States. Clin Infect Dis 2021; 73:1089-1096. [PMID: 34157096 PMCID: PMC8442791 DOI: 10.1093/cid/ciab272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-exposed, uninfected (HEU) infants experience higher rates of morbidity and mortality than HIV-unexposed, uninfected (HUU) infants. Few studies have examined whether particular infections and/or immune responses are associated with hospitalization among HEU infants born in the United States. METHODS We evaluated a subset of HEU infants enrolled in the International Maternal Pediatric Adolescent AIDS Clinical Trials Group P1025 and/or Pediatric HIV/AIDS Cohort Study Surveillance Monitoring for ART Toxicities studies. We determined seroconversion to 6 respiratory viruses and measured antibody concentrations to 9 vaccine antigens using quantitative ELISA or electrochemiluminescence. Multivariable modified Poisson regression models were fit to evaluate associations of seroconversion to each respiratory virus/family and antibody concentrations to vaccine antigens with risk of hospitalization in the first year of life. Antibody concentrations to vaccine antigens were compared between HEU infants and HUU infants from a single site using multivariable linear regression models. RESULTS Among 556 HEU infants, seroconversion to respiratory syncytial virus (RSV) and parainfluenza was associated with hospitalization (adjusted risk ratio, 1.95 [95% CI, 1.21-3.15] and 2.30 [1.42-3.73], respectively). Antibody concentrations to tetanus toxoid, pertussis, and pneumococcal vaccine antigens were higher among 525 HEU compared with 100 HUU infants. No associations were observed between antibody concentrations with any vaccine and hospitalization among HEU infants. CONCLUSIONS RSV and parainfluenza contribute to hospitalization among HEU infants in the first year of life. HEU infants demonstrate robust antibody responses to vaccine antigens; therefore, humoral immune defects likely do not explain the increased susceptibility to infection observed in this population.
Collapse
Affiliation(s)
- Christiana Smith
- Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Yanling Huo
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kunjal Patel
- Department of Epidemiology, Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kirk Fetters
- Department of Medicine, Harbor–UCLA Medical Center, Torrance, California, USA
| | - Shannon Hegemann
- College of Medicine, University of Nebraska, Omaha, Nebraska, USA
| | - Sandra Burchett
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Russell Van Dyke
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Adriana Weinberg
- Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
- Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Department of Pathology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
19
|
Links between Infections, Lung Cancer, and the Immune System. Int J Mol Sci 2021; 22:ijms22179394. [PMID: 34502312 PMCID: PMC8431665 DOI: 10.3390/ijms22179394] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the leading disease of cancer-related deaths worldwide. Since the beginning of the 20th century, various infectious agents associated with lung cancer have been identified. The mechanisms that include systemic inflammatory pathways as effect of microbial persistence in the lung can secondarily promote the development of lung carcinogenesis. Chronic inflammation associated with lung-cancer infections is known to precede tumor development, and it has a strong effect on the response(s) to therapy. In fact, both viral and bacterial infections can activate inflammatory cells and inflammatory signaling pathways. In this review, an overview of critical findings of recent studies investigating associations between each of viral and bacterial pathogens and lung carcinoma is provided, with particular emphasis on how infectious organisms can interfere with oncogenic processes and all the way through immunity. Moreover, a discussion of the direct crosstalk between lung tumor development and inflammatory processes is also presented.
Collapse
|
20
|
Baroncelli S, Galluzzo CM, Liotta G, Andreotti M, Orlando S, Ciccacci F, Mphwere R, Luhanga R, Sagno JB, Amici R, Marazzi MC, Giuliano M. HIV-exposed infants with EBV infection have a reduced persistence of the immune response to the HBV vaccine. AIDS Res Ther 2021; 18:48. [PMID: 34348748 PMCID: PMC8336389 DOI: 10.1186/s12981-021-00375-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/28/2021] [Indexed: 11/29/2022] Open
Abstract
Background In sub-Saharan African countries Epstein Barr virus (EBV) infection occurs in early childhood. We aim to investigate the factors associated with EBV acquisition and the impact of EBV infection on the humoral response to HBV vaccination in infants born from HIV-positive, antiretroviral-treated mothers in Malawi. Methods A total of 149 HIV-exposed infants were included in this longitudinal study. EBV anti-VCA IgG were measured using an ELISA assay. The EBV seroconversion was correlated with the maternal viro-immunological conditions, with infant growth and immunological vulnerability, and with the humoral response to the HBV vaccine. Results No infant was EBV-positive at 6 months (n. 52 tested). More than a third of infants (49/115 or 42.6 %) on study beyond 6 months seroconverted at 12 months. At 24 months, out of 66 tested infants, only 13 remained EBV-uninfected, while 53 (80.3 %) acquired EBV infection, rising the total proportion of EBV seroconversion to 88.7 % (102/115 infants). EBV seroconversion was significantly associated with a low maternal educational status but had no impact on infant growth or vulnerability to infections. Reduced HBsAb levels and accelerated waning of antibodies were associated with early EBV seroconversion. Conclusions We found a heterogeneous timing of acquisition of EBV with the majority of infants born from HIV + mothers acquiring infection after 6 months. Anti-HBs levels were lower and appeared to wane faster in infants acquiring EBV infection.
Collapse
|
21
|
Montoya-Ferrer A, Sanosyan A, Fayd'herbe de Maudave A, Pisoni A, Bollore K, Molès JP, Peries M, Tylleskar T, Tumwine JK, Ndeezi G, Gorgolas M, Nagot N, van de Perre P, Tuaillon E. Clinical and Biological Factors Associated With Early Epstein-Barr Virus Infection in Human Immunodeficiency Virus-Exposed Uninfected Infants in Eastern Uganda. Clin Infect Dis 2021; 72:1026-1032. [PMID: 32067040 DOI: 10.1093/cid/ciaa161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/13/2020] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Immune control of Epstein-Barr virus (EBV) infection is impaired in individuals with HIV. We explored maternal factors associated with EBV acquisition in HIV-exposed uninfected (HEU) infants and the relationship between EBV infection and serious adverse events (SAEs) during the first year of life. METHODS 201 HEU infants from Uganda enrolled in the ANRS 12174 trial were tested for antiviral capsid antigen (anti-VCA) antibodies at week 50. Date of infection was estimated by testing EBV DNA at weeks 1, 6, 14, 26, 38, and 50 postpartum on dried blood spots. RESULTS Eighty-seven (43%) infants tested positive for anti-VCA IgG at week 50. Among the 59 infants positive for EBV DNA, 25% were infected within the first 26 weeks. Almost half (12%) were infected before week 14. Shedding of EBV in breast milk was associated with EBV DNA in maternal plasma (P = .009), HIV RNA detection (P = .039), and lower CD4 count (P = .001) and correlated with plasma EBV DNA levels (P = .002). EBV infant infection at week 50 was associated with shedding of EBV in breast milk (P = .009) and young maternal age (P = .029). Occurrence of a clinical SAE, including malaria and pneumonia, was associated with higher levels of EBV DNA in infants (P = .010). CONCLUSIONS By assessing EBV infection in HEU infants we observed that infection during the first year is determined by HIV and EBV maternal factors and that EBV DNA levels were higher among infants with clinical SAEs. CLINICAL TRIALS REGISTRATION NCT00640263.
Collapse
Affiliation(s)
- Ana Montoya-Ferrer
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | - Armen Sanosyan
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | - Alexis Fayd'herbe de Maudave
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | - Amandine Pisoni
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | - Karine Bollore
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | - Jean-Pierre Molès
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | - Marianne Peries
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | | | - James K Tumwine
- Department of Pediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Grace Ndeezi
- Department of Pediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Miguel Gorgolas
- Department of Infectious Diseases, Hospital Fundación Jiménez-Díaz, University Autonoma, Madrid, Spain
| | - Nicolas Nagot
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | - Philippe van de Perre
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| | - Edouard Tuaillon
- Pathogenesis and Control of Chronic Infections, Institut national de la santé et de la recherche médicale (INSERM), Etablissement Français du Sang, University of Montpellier, Montpellier University Hospital CHU, Montpellier, Montpellier, France
| |
Collapse
|
22
|
Dried blood spots for the quantitative evaluation of IgG isotypes and correlation with serum samples in HIV-exposed uninfected (HEU) infants. J Immunol Methods 2021; 493:113019. [PMID: 33705735 DOI: 10.1016/j.jim.2021.113019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND The determination of IgG levels and their subclasses can provide clinically relevant information on the status of the immune system. Here we determined the sensitivity and reproducibility of the quantification of IgG subclasses from Dried Blood Spots (DBS) in Malawian uninfected infants exposed to HIV (HEU). METHODS Sixty paired samples of serum and DBS from HEU infants were used. Samples were collected from 1, 6, and 24-month old infants. IgGs concentrations from both serum and DBS were analyzed by BN ProSpec Siemens assay, using a different setting for sample dilutions. The reproducibility of the DBS method was tested on 10 samples run twice, starting from the DBS extraction process. To assess the systematic, proportional, and random differences, we computed the Passing-Bablok regression, and the Bland-Altman analysis to estimate the total mean bias between the two tests. RESULTS The IgG isotypes concentrations from serum and DBS showed significant differences in all the comparisons. Generally, the DBS method underestimated IgG subclasses' values showing a recovery range between 51.2% and 77.6%. Passing Bablok regression on age-based groups showed agreement for IgG, IgG1, and IgG2, but not for IgG3 and IgG4. The mean bias obtained with the Bland Altman test varied largely depending on IgG isotypes (-0.02-2.21 g/l) Coefficient of variation <7.0% was found in the repeated tests for IgG, IgG1, IgG3, and IgG4, while it was 12.4% for IgG2. CONCLUSIONS Varying degrees of differences were seen in the IgGs measurement in the two different matrices. In IgGs analysis, the DBS method offers promise for population-based research, but the results should be carefully evaluated and considered as a relative value since they are not equivalent to the serum concentrations.
Collapse
|
23
|
Sarkar A, Balogun K, Guzman Lenis MS, Acosta S, Mount HT, Serghides L. In utero exposure to protease inhibitor-based antiretroviral regimens delays growth and developmental milestones in mice. PLoS One 2020; 15:e0242513. [PMID: 33211746 PMCID: PMC7676697 DOI: 10.1371/journal.pone.0242513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/03/2020] [Indexed: 12/26/2022] Open
Abstract
Antiretroviral therapy (ART) in pregnancy has dramatically reduced HIV vertical transmission rates. Consequently, there is a growing number of children that are HIV exposed uninfected (CHEUs). Studies suggest that CHEUs exposed in utero to ART may experience developmental delays compared to their peers. We investigated the effects of in utero ART exposure on perinatal neurodevelopment in mice, through assessment of developmental milestones. Developmental milestone tests (parallel to reflex testing in human infants) are reflective of brain maturity and useful in predicting later behavioral outcomes. We hypothesized that ART in pregnancy alters the in utero environment and thereby alters developmental milestone outcomes in pups. Throughout pregnancy, dams were treated with boosted-atazanavir combined with either abacavir/lamivudine (ATV/r/ABC/3TC), or tenofovir/emtricitabine (ATV/r/TDF/FTC), or water as control. Pups were assessed daily for general somatic growth and on a battery of tests for primitive reflexes including surface-righting, negative-geotaxis, cliff-aversion, rooting, ear-twitch, auditory-reflex, forelimb-grasp, air-righting, behaviors in the neonatal open field, and olfactory test. In utero exposure to either ART regimen delayed somatic growth in offspring and evoked significant delays in the development of negative geotaxis, cliff-aversion, and ear-twitch reflexes. Exposure to ATV/r/ABC/3TC was also associated with olfactory deficits in male and forelimb grasp deficits in female pups. To explore whether delays persisted into adulthood we assessed performance in the open field test. We observed no significant differences between treatment arm for males. In females, ATV/r/TDF/FTC exposure was associated with lower total distance travelled and less ambulatory time in the centre, while ATV/r/ABC/3TC exposure was associated with higher resting times compared to controls. In utero PI-based ART exposure delays the appearance of primitive reflexes that involve vestibular and sensory-motor pathways in a mouse model. Our findings suggest that ART could be disrupting the normal progress/maturation of the underlying neurocircuits and encourage further investigation for underlying mechanisms.
Collapse
Affiliation(s)
- Ambalika Sarkar
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kayode Balogun
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Monica S. Guzman Lenis
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Sebastian Acosta
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Howard T. Mount
- Departments of Psychiatry & Physiology, Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Lena Serghides
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Women’s College Research Institute, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
24
|
Chadwick EG, Ezeanolue EE. Evaluation and Management of the Infant Exposed to HIV in the United States. Pediatrics 2020; 146:peds.2020-029058. [PMID: 33077537 DOI: 10.1542/peds.2020-029058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pediatricians play a crucial role in optimizing the prevention of perinatal transmission of HIV infection. Pediatricians provide antiretroviral prophylaxis to infants born to women with HIV type 1 (HIV) infection during pregnancy and to those whose mother's status was first identified during labor or delivery. Infants whose mothers have an undetermined HIV status should be tested for HIV infection within the boundaries of state laws and receive presumptive HIV therapy if the results are positive. Pediatricians promote avoidance of postnatal HIV transmission by advising mothers with HIV not to breastfeed. Pediatricians test the infant exposed to HIV for determination of HIV infection and monitor possible short- and long-term toxicity from antiretroviral exposure. Finally, pediatricians support families living with HIV by providing counseling to parents or caregivers as an important component of care.
Collapse
Affiliation(s)
- Ellen Gould Chadwick
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois;
| | - Echezona Edozie Ezeanolue
- HealthySunrise Foundation, Las Vegas, Nevada; and.,Department of Pediatrics, College of Medicine, University of Nigeria, Nsukka, Nigeria
| | | |
Collapse
|
25
|
Moncunill G, Dobaño C, González R, Smolen KK, Manaca MN, Balcells R, Jairoce C, Cisteró P, Vala A, Sevene E, Rupérez M, Aponte JJ, Macete E, Menéndez C, Kollmann TR, Mayor A. Association of Maternal Factors and HIV Infection With Innate Cytokine Responses of Delivering Mothers and Newborns in Mozambique. Front Microbiol 2020; 11:1452. [PMID: 32765436 PMCID: PMC7381182 DOI: 10.3389/fmicb.2020.01452] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 06/04/2020] [Indexed: 12/24/2022] Open
Abstract
Maternal factors and exposure to pathogens have an impact on infant health. For instance, HIV exposed but uninfected infants have higher morbidity and mortality than HIV unexposed infants. Innate responses are the first line of defense and orchestrate the subsequent adaptive immune response and are especially relevant in newborns. To determine the association of maternal HIV infection with maternal and newborn innate immunity we analyzed the cytokine responses upon pattern recognition receptor (PRR) stimulations in the triad of maternal peripheral and placental blood as well as in cord blood in a cohort of mother-infant pairs from southern Mozambique. A total of 48 women (35 HIV-uninfected and 13 HIV-infected) were included. Women and infant innate responses positively correlated with each other. Age, gravidity and sex of the fetus had some associations with spontaneous production of cytokines in the maternal peripheral blood. HIV-infected women not receiving antiretroviral therapy (ART) before pregnancy showed decreased IL-8 and IL-6 PRR responses in peripheral blood compared to those HIV-uninfected, and PRR hyporesponsiveness for IL-8 was also found in the corresponding infant’s cord blood. HIV infection had a greater impact on placental blood responses, with significantly increased pro-inflammatory, TH1 and TH17 PRR responses in HIV-infected women not receiving ART before pregnancy compared to HIV-uninfected women. In conclusion, innate response of the mother and her newborn was altered by HIV infection in the women who did not receive ART before pregnancy. As these responses could be related to birth outcomes, targeted innate immune modulation could improve maternal and newborn health.
Collapse
Affiliation(s)
- Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel González
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Department of Experimental Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Kinga K Smolen
- Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Maria N Manaca
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Reyes Balcells
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Chenjerai Jairoce
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Pau Cisteró
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Anifa Vala
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Esperança Sevene
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Department of Physiological Science, Clinical Pharmacology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - María Rupérez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - John J Aponte
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Eusébio Macete
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Clara Menéndez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Tobias R Kollmann
- Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
26
|
Labuda SM, Huo Y, Kacanek D, Patel K, Huybrechts K, Jao J, Smith C, Hernandez-Diaz S, Scott G, Burchett S, Kakkar F, Chadwick EG, Van Dyke RB. Rates of Hospitalization and Infection-Related Hospitalization Among Human Immunodeficiency Virus (HIV)-Exposed Uninfected Children Compared to HIV-Unexposed Uninfected Children in the United States, 2007-2016. Clin Infect Dis 2020; 71:332-339. [PMID: 31504291 PMCID: PMC7353328 DOI: 10.1093/cid/ciz820] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/19/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Studies from multiple countries have suggested impaired immunity in perinatally human immunodeficiency virus (HIV)-exposed uninfected children (HEU), with elevated rates of all-cause hospitalization and infections. We estimated and compared the incidence of all-cause hospitalization and infection-related hospitalization in the first 2 years of life among HEU and HIV-unexposed uninfected children (HUU) in the United States. Among HEU, we evaluated associations of maternal HIV disease-related factors during pregnancy with risk of child hospitalization. METHODS HEU data from subjects enrolled in the Surveillance Monitoring for Antiretroviral Therapy Toxicities Study (SMARTT) cohort who were born during 2006-2017 were analyzed. HUU comparison data were obtained from the Medicaid Analytic Extract database, restricted to states participating in SMARTT. We compared rates of first hospitalization, total hospitalizations, first infection-related hospitalization, total infection-related hospitalizations, and mortality between HEU and HUU using Poisson regression. Among HEU, multivariable Poisson regression models were fitted to evaluate associations of maternal HIV factors with risk of hospitalization. RESULTS A total of 2404 HEU and 3 605 864 HUU were included in the analysis. HEU children had approximately 2 times greater rates of first hospitalization, total hospitalizations, first infection-related hospitalization, and total infection-related hospitalizations compared with HUUs. There was no significant difference in mortality. Maternal HIV disease factors were not associated with the risk of child infection or hospitalization. CONCLUSIONS Compared with HUU, HEU children in the United States have higher rates of hospitalization and infection-related hospitalization in the first 2 years of life, consistent with studies in other countries. Closer monitoring of HEU infants for infection and further elucidation of immune mechanisms is needed.
Collapse
Affiliation(s)
- Sarah M Labuda
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yanling Huo
- Center for Biostatistics in AIDS Research,Boston, Massachusetts, USA
| | - Deborah Kacanek
- Center for Biostatistics in AIDS Research,Boston, Massachusetts, USA
| | - Kunjal Patel
- Center for Biostatistics in AIDS Research,Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Krista Huybrechts
- Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Jao
- Department of Pediatrics, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christiana Smith
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Gwendolyn Scott
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sandra Burchett
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Fatima Kakkar
- Department of Pediatrics, Université de Montréal, Montreal, Canada
| | - Ellen G Chadwick
- Department of Pediatrics, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Russell B Van Dyke
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
27
|
Ajaykumar A, Zhu M, Kakkar F, Brophy J, Bitnun A, Alimenti A, Soudeyns H, Saberi S, Albert AYK, Money DM, Côté HCF. Elevated Blood Mitochondrial DNA in Early Life Among Uninfected Children Exposed to Human Immunodeficiency Virus and Combination Antiretroviral Therapy in utero. J Infect Dis 2020; 223:621-631. [PMID: 32638023 DOI: 10.1093/infdis/jiaa410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/03/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Combination antiretroviral therapy (cART) during pregnancy prevents vertical transmission, but many antiretrovirals cross the placenta and several can affect mitochondria. Exposure to maternal human immunodeficiency virus (HIV) and/or cART could have long-term effects on children who are HIV exposed and uninfected (CHEU). Our objective was to compare blood mitochondrial DNA (mtDNA) content in CHEU and children who are HIV unexposed and uninfected (CHUU), at birth and in early life. METHODS Whole-blood mtDNA content at birth and in early life (age 0-3 years) was compared cross-sectionally between CHEU and CHUU. Longitudinal changes in mtDNA content among CHEU was also evaluated. RESULTS At birth, CHEU status and younger gestational age were associated with higher mtDNA content. These remained independently associated with mtDNA content in multivariable analyses, whether considering all infants, or only those born at term. Longitudinally, CHEU mtDNA levels remained unchanged during the first 6 months of life, and gradually declined thereafter. A separate age- and sex-matched cross-sectional analysis (in 214 CHEU and 214 CHUU) illustrates that the difference in mtDNA between the groups remains detectable throughout the first 3 years of life. CONCLUSION The persistently elevated blood mtDNA content observed among CHEU represents a long-term effect, possibly resulting from in utero stresses related to maternal HIV and/or cART. The clinical impact of altered mtDNA levels is unclear.
Collapse
Affiliation(s)
- Abhinav Ajaykumar
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mayanne Zhu
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fatima Kakkar
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Jason Brophy
- Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - Ari Bitnun
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Ariane Alimenti
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,BC Women's Hospital and Health Centre, Vancouver, British Columbia, Canada
| | - Hugo Soudeyns
- Unité d'Immunopathologie Virale, Centre de Recherche du CHU Sainte-Justine, Montreal, Quebec, Canada.,Department of Microbiology, Infectiology & Immunology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Sara Saberi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Deborah M Money
- BC Women's Hospital and Health Centre, Vancouver, British Columbia, Canada.,Women's Health Research Institute, Vancouver, British Columbia, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hélène C F Côté
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada.,Women's Health Research Institute, Vancouver, British Columbia, Canada
| | | |
Collapse
|
28
|
Huq F, Obida M, Bornman R, Di Lenardo T, Chevrier J. Associations between prenatal exposure to DDT and DDE and allergy symptoms and diagnoses in the Venda Health Examination of Mothers, Babies and their Environment (VHEMBE), South Africa. ENVIRONMENTAL RESEARCH 2020; 185:109366. [PMID: 32299029 PMCID: PMC7336873 DOI: 10.1016/j.envres.2020.109366] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 06/11/2023]
Abstract
Dichlorodiphenyl trichloroethane (DDT) is an organochlorine insecticide that is banned internationally except for use as part of Indoor Residual Spraying (IRS) programs to control malaria. Although animal studies show that DDT and its breakdown product dichlorodiphenyl dichloroethylene (DDE) affect the immune system and may cause allergies, no studies have examined this question in populations where IRS is conducted. The aim of our study was to investigate whether prenatal exposure to DDT and DDE is associated with allergy symptoms and diagnose among South African children living in an area where IRS is conducted. To accomplish this aim, we used data from the Venda Health Examination of Mothers, Babies and their Environment (VHEMBE), an ongoing birth cohort study of 752 children born between 2012 and 2013 in the rural Vhembe district of Limpopo, South Africa. We measured maternal peripartum serum concentrations of DDT and DDE, and administered a questionnaire to the caregivers of 658 children aged 3.5 years to collect information on allergy symptoms and diagnoses as well as potential confounders using validated instruments. Using multiple logistic regression models, we found positive associations between DDT and DDE serum concentrations and most of the allergy symptoms and diagnoses. Maternal DDT (Odds Ratio [OR] = 1.5 per 10-fold increase, 95% Confidence interval, CI = 1.0, 2.3) and DDE (OR = 1.4, 95% CI = 0.8, 2.4) serum concentrations were most strongly associated with caregiver report of wheezing or whistling in the chest. Concentrations of DDT and/or DDE were also associated with increased odds of children's chests sounding wheezy during or after exercise, itchy rashes coming and going for at least six months, diagnosis of food allergy, and diagnosis of dust or dust mites allergy but confidence intervals crossed the null. Results suggest that prenatal exposure to DDT, and possibly DDE, is associated with elevated odds of wheezing among children from an IRS area.
Collapse
Affiliation(s)
- Fahmida Huq
- Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Muvhulawa Obida
- University of Pretoria School of Health Systems and Public Health, and Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Riana Bornman
- University of Pretoria School of Health Systems and Public Health, and Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Thomas Di Lenardo
- Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Jonathan Chevrier
- Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montréal, QC, Canada.
| |
Collapse
|
29
|
Eckard AR, Kirk SE, Hagood NL. Contemporary Issues in Pregnancy (and Offspring) in the Current HIV Era. Curr HIV/AIDS Rep 2020; 16:492-500. [PMID: 31630334 DOI: 10.1007/s11904-019-00465-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Although antiretroviral therapy (ART) has dramatically reduced mother to child transmission of HIV, data continue to mount that infants exposed to HIV in utero but are not infected (HEU) have serious negative health consequences compared to unexposed infants. This review evaluates recent literature on contemporary issues related to complications seen in pregnant women with HIV and their offspring. RECENT FINDINGS Current studies show that HEU infants are at a high risk of adverse outcomes, including premature birth, poor growth, neurodevelopmental impairment, immune dysfunction, infectious morbidity, and death. Etiologies for the observed clinical events and subclinical alterations are complex and multifactorial, and the long-term consequences of many findings are yet unknown. HEU infants have an unacceptable rate of morbidity and mortality from perinatal HIV and ART exposure, even in the modern ART era. Continual monitoring and reporting is imperative to protect this vulnerable population in our everchanging landscape of HIV treatment and prevention.
Collapse
Affiliation(s)
- Allison Ross Eckard
- Departments of Pediatrics and Medicine, Divisions of Infectious Diseases, Medical University of South Carolina, 135 Rutledge Ave., MSC 752, Charleston, SC, USA.
| | - Stephanie E Kirk
- Departments of Pediatrics and Medicine, Divisions of Infectious Diseases, Medical University of South Carolina, 135 Rutledge Ave., MSC 752, Charleston, SC, USA
| | - Nancy L Hagood
- Departments of Pediatrics and Medicine, Divisions of Infectious Diseases, Medical University of South Carolina, 135 Rutledge Ave., MSC 752, Charleston, SC, USA
| |
Collapse
|
30
|
Baroncelli S, Galluzzo CM, Liotta G, Andreotti M, Orlando S, Ciccacci F, Jere H, Luhanga R, Sagno JB, Amici R, Marazzi MC, Giuliano M. Dynamics of immunoglobulin G subclasses during the first two years of life in Malawian infants born to HIV-positive mothers. BMC Pediatr 2020; 20:181. [PMID: 32326903 PMCID: PMC7178742 DOI: 10.1186/s12887-020-02091-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/16/2020] [Indexed: 11/24/2022] Open
Abstract
Background Maternal antibodies are key components of the protective responses of infants who are unable to produce their own IgG until 6 months of life. There is evidence that HIV-exposed uninfected children (HEU) have IgG levels abnormalities, that can be partially responsible for the higher vulnerability to infections in the first 2 years of the life of this population. This retrospective study aimed to characterize the dynamics in plasma levels of total IgG and their isotypes during the first 2 years of life in HEU infants exclusively breastfed through 6 months of age. Methods Total IgG, IgG1, IgG2, IgG3 and IgG4 isotypes, and IgM and IgA plasma concentrations were determined by nephelometric methods in 30 Malawian infants born to HIV-positive women at month 1, 6 and 24 of life. Results At 1-month infants had a median concentration of total IgG of 8.48 g/l, (IQR 7.57–9.15), with an overrepresentation of the IgG1 isotype (89.0% of total) and low levels of IgG2 (0.52 g/l, IQR, 0.46–0.65). Total IgG and IgG1 concentrations were lower at 6 months (− 2.1 and − 1.12 g/dl, respectively) reflecting disappearance of maternal antibodies, but at 24 months their levels were higher with respect to the reported reference values for age-matched pairs. Abnormal isotype distribution was still present at 24 months with IgG2 remaining strongly underrepresented (0.87 g/l, 7.5% of total IgG). Conclusion HIV exposure during pregnancy and breastfeeding seems to influence the IgG maturation and isotype distribution that persist in 2-year old infants.
Collapse
Affiliation(s)
- Silvia Baroncelli
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Clementina Maria Galluzzo
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Giuseppe Liotta
- Department of Biomedicine and Preventio, University of Rome Tor Vergata, Rome, Italy
| | - Mauro Andreotti
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Stefano Orlando
- Department of Biomedicine and Preventio, University of Rome Tor Vergata, Rome, Italy
| | - Fausto Ciccacci
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Haswell Jere
- DREAM Program, Community of S. Egidio, Blantyre, Malawi
| | | | | | - Roberta Amici
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | | | - Marina Giuliano
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
31
|
Greiter BM, Kahlert CR, Eberhard N, Sultan-Beyer L, Berger C, Paioni P. Lymphocyte Subsets in HIV-Exposed Uninfected Infants: The Impact of Neonatal Postexposure Prophylaxis With Zidovudine. Open Forum Infect Dis 2020; 7:ofaa108. [PMID: 32368562 PMCID: PMC7190205 DOI: 10.1093/ofid/ofaa108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/26/2020] [Indexed: 11/13/2022] Open
Abstract
HIV-exposed, uninfected (HEU) infants receiving neonatal postexposure prophylaxis with zidovudine showed nonsignificant trends of lower CD4 and CD8 T cells as well as CD19 B cells than those who did not, suggesting toxicity that might impact the overall health of HEU children.
Collapse
Affiliation(s)
- Beat M Greiter
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Christian R Kahlert
- Children's Hospital of Eastern Switzerland, Infectious Diseases and Hospital Epidemiology, St. Gallen, Switzerland.,Cantonal Hospital St. Gallen, Infectious Diseases and Hospital Epidemiology, St. Gallen, Switzerland
| | - Nadia Eberhard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Leila Sultan-Beyer
- Department of Gynaecology, University Hospital Zurich, Zurich, Switzerland
| | - Christoph Berger
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Paolo Paioni
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Smith C, Moraka NO, Ibrahim M, Moyo S, Mayondi G, Kammerer B, Leidner J, Gaseitsiwe S, Li S, Shapiro R, Lockman S, Weinberg A. Human Immunodeficiency Virus Exposure but Not Early Cytomegalovirus Infection Is Associated With Increased Hospitalization and Decreased Memory T-Cell Responses to Tetanus Vaccine. J Infect Dis 2020; 221:1167-1175. [PMID: 31711179 PMCID: PMC7075416 DOI: 10.1093/infdis/jiz590] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/07/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-exposed, uninfected (HEU) infants experience high rates of infectious morbidity. We hypothesized that early cytomegalovirus (CMV) infection was associated with increased hospitalization rates and decreased vaccine responses in HEU compared with HIV-unexposed (HUU) infants. METHODS Among infants enrolled in the Tshipidi study in Botswana, we determined CMV infection status by 6 months of age and compared hospitalization rates and responses to tetanus and Bacille Calmette-Guérin vaccines among HEU and HUU vaccinees. RESULTS Fifteen of 226 (6.6%) HEU infants and 17 (19.3%) of 88 HUU infants were CMV-infected by 6 months. The HEU infants were approximately 3 times as likely to be hospitalized compared with HUU infants (P = .02). The HEU peripheral blood cells produced less interleukin (IL)-2 (P = .004), but similar amounts of interferon-γ, after stimulation with tetanus toxoid. Antitetanus immunoglobulin G titers were similar between groups. Cellular responses to purified protein derivative stimulation did not differ between groups. Maternal receipt of 3-drug antiretroviral therapy compared with zidovudine was associated with increased IL-2 expression after tetanus toxoid stimulation. The infants' CMV infection status was not associated with clinical or vaccine response outcomes. CONCLUSIONS We observed that increased rates of hospitalization and decreased memory T-cell responses to tetanus vaccine were associated with HIV exposure and incomplete treatment of maternal HIV infection, but not early CMV infection.
Collapse
Affiliation(s)
- Christiana Smith
- University of Colorado School of Medicine, Denver, Colorado, USA
| | - Natasha O Moraka
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Stellenbosch University, Stellenbosch, South Africa
| | | | - Sikhulile Moyo
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Gloria Mayondi
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Betsy Kammerer
- Boston Children’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Jean Leidner
- Goodtables Data Consulting, Norman, Oklahoma, USA
| | - Simani Gaseitsiwe
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Shaobing Li
- University of Colorado School of Medicine, Denver, Colorado, USA
| | - Roger Shapiro
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Shahin Lockman
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Adriana Weinberg
- University of Colorado School of Medicine, Denver, Colorado, USA
| |
Collapse
|
33
|
Immunobiological aspects of vaccines in pregnancy: Maternal perspective. MATERNAL IMMUNIZATION 2020. [PMCID: PMC7149477 DOI: 10.1016/b978-0-12-814582-1.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunization during pregnancy is an efficient strategy to protect both the mother and the newborn infant against infectious pathogens. Pregnant women have an increased susceptibility to severe infections caused by some pathogens, but the mechanisms involved remain poorly understood. Pregnancy is associated with dynamic changes in maternal immune system that are critical for tolerance of the fetus. These changes could also play an important role in shaping maternal immune components that are transferred to the newborn infant following natural infection or vaccination to prevent infectious diseases in early life. As the momentum for maternal immunization is growing, there is a need to increase our understanding of the immunobiology of maternal immunization in order to better prevent infectious diseases in the pregnant women and the young infant.
Collapse
|
34
|
Bobat R. Opportunistic Infections. HIV INFECTION IN CHILDREN AND ADOLESCENTS 2020. [PMCID: PMC7120925 DOI: 10.1007/978-3-030-35433-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Evolution in paediatric HIV management has changed the incidence and prevalence of opportunistic infections and a major reduction has been shown for most opportunistic infections with antiretroviral therapy use in lower and middle-income countries, especially in the first year of treatment. However, the high prevalence of disease still requires adequate management of opportunistic infections, to improve patient quality of life and the impact on burden of disease. Lower CD4 counts were associated with chronic infection and increased risk of opportunistic infections in patients, but some studies have shown that even children with high CD4 counts may have opportunistic infections. This chapter reviews common opportunistic infections that may infect HIV positive children and adolescents, particularly in sub Saharan Africa.
Collapse
Affiliation(s)
- Raziya Bobat
- Department of Paediatrics and Child Health, University of KwaZulu-Natal, Durban, KwaZulu-Natal South Africa
| |
Collapse
|
35
|
Merino KM, Slisarenko N, Taylor JM, Falkenstein KP, Gilbert MH, Bohm RP, Blanchard JL, Ardeshir A, Didier ES, Kim WK, Kuroda MJ. Clinical and Immunological Metrics During Pediatric Rhesus Macaque Development. Front Pediatr 2020; 8:388. [PMID: 32766187 PMCID: PMC7378395 DOI: 10.3389/fped.2020.00388] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Clinical measurements commonly used to evaluate overall health of laboratory animals including complete blood count, serum chemistry, weight, and immunophenotyping, differ with respect to age, development, and environment. This report provides comprehensive clinical and immunological reference ranges for pediatric rhesus macaques over the first year of life. Methods: We collected and analyzed blood samples from 151 healthy rhesus macaques, aged 0-55 weeks, and compared mother-reared infants to two categories of nursery-reared infants; those on an active research protocol and those under derivation for the expanded specific-pathogen-free breeding colony. Hematology was performed on EDTA-anticoagulated blood using a Sysmex XT2000i, and serum clinical chemistry was performed using the Beckman AU480 chemistry analyzer. Immunophenotyping of whole blood was performed with immunofluorescence staining and subsequent flow cytometric analysis on a BD LSRFortessa. Plasma cytokine analysis was performed using a Millipore multiplex Luminex assay. Results: For hematological and chemistry measurements, pediatric reference ranges deviate largely from adults. Comparison of mother-reared and nursery-reared animals revealed that large differences depend on rearing conditions and diet. Significant differences found between two nursery-reared cohorts (research and colony animals) indicate large influences of experimental factors and anesthetic events on these parameters. Immune cells and cytokine responses presented with distinct patterns for infants depending on age, birth location, and rearing conditions. Conclusions: Our results illustrate how the immune system changed over time and that there was variability among pediatric age groups. Reference ranges of results reported here will support interpretations for how infection and treatment may skew common immune correlates used for assessment of pathology or protection in research studies as well as help veterinarians in the clinical care of infant non-human primates. We highlighted the importance of using age-specific reference comparisons for pediatric studies and reiterated the utility of rhesus macaques as a model for human studies. Given the rapid transformation that occurs in multiple tissue compartments after birth and cumulative exposures to antigens as individuals grow, a better understanding of immunological development and how this relates to timing of infection or vaccination will support optimal experimental designs for developing vaccines and treatment interventions.
Collapse
Affiliation(s)
- Kristen M Merino
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, United States.,Walter Reed Army Institute of Research, National Academy of Sciences, Engineering and Medicine Fellow, Silver Spring, MD, United States
| | - Nadia Slisarenko
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, United States
| | - Joshua M Taylor
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Kathrine P Falkenstein
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Margaret H Gilbert
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Rudolf P Bohm
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - James L Blanchard
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Amir Ardeshir
- California National Primate Research Center, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Elizabeth S Didier
- Center for Immunology and Infectious Diseases, California National Primate Research Center, University of California, Davis, Davis, CA, United States.,Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Marcelo J Kuroda
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, United States.,Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| |
Collapse
|
36
|
Musimbi ZD, Rono MK, Otieno JR, Kibinge N, Ochola-Oyier LI, de Villiers EP, Nduati EW. Peripheral blood mononuclear cell transcriptomes reveal an over-representation of down-regulated genes associated with immunity in HIV-exposed uninfected infants. Sci Rep 2019; 9:18124. [PMID: 31792230 PMCID: PMC6889308 DOI: 10.1038/s41598-019-54083-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022] Open
Abstract
HIV-exposed uninfected (HEU) infants are disproportionately at a higher risk of morbidity and mortality, as compared to HIV-unexposed uninfected (HUU) infants. Here, we used transcriptional profiling of peripheral blood mononuclear cells to determine immunological signatures of in utero HIV exposure. We identified 262 differentially expressed genes (DEGs) in HEU compared to HUU infants. Weighted gene co-expression network analysis (WGCNA) identified six modules that had significant associations with clinical traits. Functional enrichment analysis on both DEGs and the six significantly associated modules revealed an enrichment of G-protein coupled receptors and the immune system, specifically affecting neutrophil function and antibacterial responses. Additionally, malaria pathogenicity genes (thrombospondin 1-(THBS 1), interleukin 6 (IL6), and arginine decarboxylase 2 (ADC2)) were down-regulated. Of interest, the down-regulated immunity genes were positively correlated to the expression of epigenetic factors of the histone family and high-mobility group protein B2 (HMGB2), suggesting their role in the dysregulation of the HEU transcriptional landscape. Overall, we show that genes primarily associated with neutrophil mediated immunity were repressed in the HEU infants. Our results suggest that this could be a contributing factor to the increased susceptibility to bacterial infections associated with higher morbidity and mortality commonly reported in HEU infants.
Collapse
Affiliation(s)
- Zaneta D Musimbi
- Center of Biotechnology and Bioinformatics, Chiromo Campus, University of Nairobi, Nairobi, Kenya.
| | - Martin K Rono
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya.
| | | | | | - Lynette Isabella Ochola-Oyier
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya
| | - Etienne Pierre de Villiers
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Eunice W Nduati
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya
| |
Collapse
|
37
|
Siawaya ACM, Mveang-Nzoghe A, Mpega CNM, Leboueny M, Ndjindji OM, Ndong AM, Essone PN, Siawaya JFD. Increased platelets count in HIV-1 uninfected infants born from HIV-1 infected mothers. Hematol Rep 2019; 11:7056. [PMID: 31583065 PMCID: PMC6775485 DOI: 10.4081/hr.2019.7056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 06/06/2017] [Indexed: 12/03/2022] Open
Abstract
HIV-exposed uninfected infants (HEU) represent a growing population in developing countries including Gabon. Several studies have shown the vulnerability of these infants toward infectious diseases. The aim of the study was to contribute to the global effort to understand how HIVexposure or anti retroviral therapy affects infants’ blood elements. We assessed HEU infants’ complete blood count using a blood analyzer instrument. Our investigations showed that among the observed clinically relevant hematological abnormalities events, thrombocytosis was the most prevalent clinically relevant hematological abnormality associated with HEU infants’. We showed that HEU infants had significantly higher platelets count than HUinfants. Therefore, higher level of platelets seems to characterize HEU infants when compared to HU infants.
Collapse
Affiliation(s)
| | - Amandine Mveang-Nzoghe
- Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique, Libreville, Gabon
| | - Chérone Nancy Mbani Mpega
- Département de Chimie, Faculté des Sciences, Université des Sciences et Techniques de Masuku, Franceville, Gabon
| | - Marielle Leboueny
- Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique, Libreville, Gabon
| | - Ofilia Mvoundza Ndjindji
- Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique, Libreville, Gabon
| | - Armel Mintsa Ndong
- Unité de Virologie, Laboratoire National de Santé Publique, Libreville, Gabon
| | - Paulin N Essone
- Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique, Libreville, Gabon.,Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Science, University of Cape Town, South Africa
| | - Joel Fleury Djoba Siawaya
- Unités de Recherche et de Diagnostics Spécialisés, Laboratoire National de Santé Publique, Libreville, Gabon
| |
Collapse
|
38
|
Dambaya B, Nkenfou CN, Ambada G, Ikomey GM, Mouafo LM, Ngoufack N, Ndzi EN, Této G, Nanfack A, Sonela N, Fokam J, Flobert N, Colizzi V, Ndjolo A. Differential expression of Fas receptors (CD95) and Fas ligands (CD95L) in HIV infected and exposed uninfected children in Cameroon versus unexposed children. Pan Afr Med J 2019; 34:39. [PMID: 31762906 PMCID: PMC6859037 DOI: 10.11604/pamj.2019.34.39.15038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/04/2019] [Indexed: 11/11/2022] Open
Abstract
Introduction The number of HIV exposed uninfected (HEU) infants is increasing as vertical transmission is reducing. This subpopulation requires more investigations. This study aimed at comparing the expression level of soluble Fas receptors (FasR) and ligands (FasL) between HIV infected, HEU and unexposed children. Methods Eighty eight HIV-1infected, 86 HEU and 38 HIV unexposed children were recruited. Soluble FasR and FasL were measured in their plasma. Mann-Whitney U-Test was used to compare groups with 95% confidence. Spearman coefficient was used to test the correlation with CD4 and viral load (VL). Results Overall plasma levels of FasR were higher than that of FasL. The concentration of FasR and FasL were significantly higher in HIV-1 infected children in comparison to HEU and unexposed children. There was no difference in the plasma level of FasL in HIV infected compared to HEU children. A significant difference was observed between HIV infected children and HEU children (P=0.001) for the FasL. FasR were higher in both HIV infected and unexposed children compared to HEU children. There was a positive correlation (rs=+0.4; p=0.01) in ARV treated children between CD4 count and FasL concentration. Significant negative correlation (rs=-0.3; p=0.040) in ARV naïve children was observed between CD4 percentage and FasL. Significant and positive correlation (rs=+0.4; p=0.008) was observed between the VL and FasL in HIV infected, treated or not. Conclusion HEU children differ from HIV infected and unexposed children as the level of FasL/R expression is concerned. HEU should be considered different from HIV unexposed although exempt from virus as some immune dysfunctions have been reported among them.
Collapse
Affiliation(s)
- Béatrice Dambaya
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon.,Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Céline Nguefeu Nkenfou
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon.,Higher Teachers' Training College, University of Yaounde I, Yaounde, Cameroon
| | - Georgia Ambada
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon.,Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | | | | | - Nicole Ngoufack
- Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | | | - Georges Této
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon
| | - Aubin Nanfack
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon
| | - Nelson Sonela
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon.,Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Joseph Fokam
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon
| | - Njiokou Flobert
- Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Vittorio Colizzi
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon.,University of Tor Vergata, Rome, Italy
| | - Alexis Ndjolo
- Chantal Biya International Reference Centre for Research on HIV/AIDS prevention and management-CBIRC, Yaounde, Cameroon.,Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde, Cameroon
| |
Collapse
|
39
|
Deichsel EL, Pavlinac PB, Richardson BA, Mbori-Ngacha D, Walson JL, McGrath CJ, Farquhar C, Bosire R, Maleche-Obimbo E, John-Stewart GC. Birth size and early pneumonia predict linear growth among HIV-exposed uninfected infants. MATERNAL AND CHILD NUTRITION 2019; 15:e12861. [PMID: 31222958 DOI: 10.1111/mcn.12861] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/27/2022]
Abstract
Stunting remains a global health priority, particularly in sub-Saharan Africa. Identifying determinants of linear growth in HIV-exposed uninfected (HEU) infants can inform interventions to prevent stunting in this vulnerable population. HIV-infected mothers and their uninfected infants were followed monthly from pregnancy to 12-month post-partum in Nairobi, Kenya. Mixed-effects models estimated the change in length-for-age z-score (LAZ) from birth to 12 months by environmental, maternal, and infant characteristics. Multivariable models included factors univariately associated with LAZ. Among 372 HEU infants, mean LAZ decreased from -0.54 (95% confidence interval [CI] [-0.67, -0.41]) to -1.09 (95% CI [-1.23, -0.96]) between 0 and 12 months. Declines in LAZ were associated with crowding (≥2 persons per room; adjusted difference [AD] in 0-12 month change: -0.46; 95% CI [-0.87, -0.05]), use of a pit latrine versus a flush toilet (AD: -0.29; 95% CI [-0.57, -0.02]), and early infant pneumonia (AD: -1.14; 95% CI [-1.99, -0.29]). Infants with low birthweight (<2,500 g; AD: 1.08; 95% CI [0.40, 1.76]) and birth stunting (AD: 1.11; 95% CI [0.45, 1.78]) experienced improved linear growth. By 12 months of age, 46 infants were stunted, of whom 11 (24%) were stunted at birth. Of the 34 infants stunted at birth with an available 12-month LAZ, 68% were not stunted at 12 months. Some low birthweight and birth-stunted HEU infants had significant linear growth recovery. Early infant pneumonia and household environment predicted poor linear growth and may identify a subgroup of HEU infants for whom to provide growth-promoting interventions.
Collapse
Affiliation(s)
- Emily L Deichsel
- Biostatistics, Global Health, Epidemiology, Medicine, University of Washington, Seattle, Washington
| | - Patricia B Pavlinac
- Biostatistics, Global Health, Epidemiology, Medicine, University of Washington, Seattle, Washington
| | - Barbra A Richardson
- Biostatistics, Global Health, Epidemiology, Medicine, University of Washington, Seattle, Washington
| | | | - Judd L Walson
- Biostatistics, Global Health, Epidemiology, Medicine, University of Washington, Seattle, Washington.,Child Acute Illness and Nutrition (CHAIN) Network, Nairobi, Kenya
| | - Christine J McGrath
- Biostatistics, Global Health, Epidemiology, Medicine, University of Washington, Seattle, Washington
| | - Carey Farquhar
- Biostatistics, Global Health, Epidemiology, Medicine, University of Washington, Seattle, Washington
| | - Rose Bosire
- Center for Public Health Research, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | | | - Grace C John-Stewart
- Biostatistics, Global Health, Epidemiology, Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
40
|
Ekali GL, Jesson J, Enok PB, Leroy V. Effect of in utero exposure to HIV and antiretroviral drugs on growth in HIV-exposed uninfected children: a systematic review and meta-analysis protocol. BMJ Open 2019; 9:e023937. [PMID: 31229997 PMCID: PMC6596996 DOI: 10.1136/bmjopen-2018-023937] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION HIV-exposed uninfected (HEU) children have higher morbidity and mortality compared with HIV unexposed uninfected children. Despite the fact that malnutrition contributes to about half of all infant deaths below 5 years of age in low-income and middle-income countries and that growth impairment has been reported in the HEU population, the spectrum of growth disorders associated with HIV and antiretroviral therapy (ART) exposure during the in utero and perinatal periods is yet to comprehensively summarised among the global HEU population. This protocol for a systematic review and meta-analysis aims to critically synthesise data concerning the prevalence of underweight, stunting and wasting at different ages in the global HEU population. METHODS AND ANALYSIS Medline, EMBASE, Cochrane Library, TOXLINE, WHO Global Index Medicus and the Web of Science will be searched for relevant articles published between 1 January 1989 and 1 December 2017 without language restriction. In addition, conference abstracts and reference lists of eligible papers and relevant review articles will be screened. Authors will screen and select studies, extract data, assess the risk of bias as well as studies individually for heterogeneity. Study-specific estimates will be pooled through a random-effects meta-analysis model for studies that are clinically homogeneous while funnel plots and Egger's test will be used to detect publication bias. Results will be presented by ART availability period, country income levels and mode of breastfeeding. ETHICS AND DISSEMINATION Ethical approval will not be required for this study because it will be based on published data. The final report of this study will be published in a peer-reviewed journal and presented at scientific conferences. This review will summarise the evidence and quantify the problem of growth impairment in HEU infants and so shed more light on our understanding of the higher morbidity and mortality in this growing population. PROSPERO REGISTRATION NUMBER CRD42018091762.
Collapse
Affiliation(s)
- Gabriel L Ekali
- Biotechnology Center, Universite de Yaounde I, Yaounde, Cameroon
- National AIDS Control Committee, Cameroon
- Inserm UMR 1027, Universite Toulouse III Paul Sabatier, Toulouse, France
| | - Julie Jesson
- Inserm UMR 1027, Universite Toulouse III Paul Sabatier, Toulouse, France
| | - Pascal B Enok
- Department of Preventive Medicine, University of Montreal, Montreal, Canada
| | | |
Collapse
|
41
|
Slogrove AL, Powis KM, Cotton MF. Human Immunodeficiency Virus-exposed Uninfected Infants: Surviving and Thriving or Overlooked by Success? Clin Infect Dis 2019; 68:2156-2158. [PMID: 30535339 PMCID: PMC6769231 DOI: 10.1093/cid/ciy1056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Amy L Slogrove
- Department of Paediatrics and Child Health, Faculty of Medicine & Health Sciences, Stellenbosch University, Worcester, South Africa
- Ukwanda Centre for Rural Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Worcester, South Africa
| | - Kathleen M Powis
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Massachusetts General Hospital, Boston
- Department of Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Mark F Cotton
- Division of Infectious Diseases and Family Clinical Research Unit, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
42
|
Goetghebuer T, Smolen KK, Adler C, Das J, McBride T, Smits G, Lecomte S, Haelterman E, Barlow P, Piedra PA, Van der Klis F, Kollmann TR, Lauffenburger DA, Alter G, Levy J, Marchant A. Reply to Slogrove et al. Clin Infect Dis 2019; 68:2158. [PMID: 30535371 DOI: 10.1093/cid/ciy1058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Kinga K Smolen
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
| | | | - Jishnu Das
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Trevor McBride
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Gaby Smits
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Sandra Lecomte
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
| | | | - Patricia Barlow
- Department of Obstetrics and Gynecology, Hôpital Saint-Pierre, Brussels, Belgium
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Fiona Van der Klis
- National Institute of Health and the Environment, Bilthoven, The Netherlands
| | - Tobias R Kollmann
- Division of Infectious Diseases, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University
| | - Jack Levy
- Department of Pediatrics, Hôpital Saint-Pierre, Brussels
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
| |
Collapse
|
43
|
Moraka NO, Moyo S, Mayondi G, Leidner J, Ibrahim M, Smith C, Weinberg A, Li S, Thami PK, Kammerer B, Ajibola G, Musonda R, Shapiro R, Gaseitsiwe S, Lockman S. Cytomegalovirus Viremia in HIV-1 Subtype C Positive Women at Delivery in Botswana and Adverse Birth/Infant Health Outcomes. J Acquir Immune Defic Syndr 2019; 81:118-124. [PMID: 30964806 PMCID: PMC7029790 DOI: 10.1097/qai.0000000000001982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We evaluated the association between maternal cytomegalovirus (CMV) viremia during pregnancy and adverse birth and infant health outcomes in HIV-infected mothers and their HIV-exposed uninfected infants. METHODS HIV-positive women and their infants were followed prospectively from pregnancy through 2 years postpartum in the "Tshipidi" study in Botswana. We analyzed the association between detectable CMV DNA in maternal blood at delivery and adverse birth outcomes (stillbirth, preterm delivery, small for gestational age, or birth defect), as well as infant hospitalization and mortality through 24 months. RESULTS We measured CMV DNA in blood samples from 350 (77.1%) of 454 HIV-positive women from the Tshipidi study. The median maternal CD4 count was 422 cells/mL, and median HIV-1 RNA at entry was 3.2 log10 copies/mL. Fifty-one (14.6%) women had detectable CMV DNA. In unadjusted analyses, detectable CMV DNA was associated with higher maternal HIV-1 RNA [odds ratio (OR) 1.4, 95% confidence interval (CI): 1.1 to 1.9], presence of a birth defect (OR 9.8, 95% CI: 1.6 to 60.3), and occurrence of any adverse birth outcome (OR 2.0, 95% CI: 1.04 to 3.95). In multivariable analysis, we observed a trend toward association between detectable maternal CMV DNA and occurrence of any adverse birth outcome (adjusted OR 1.9, 95% CI: 0.96 to 3.8). Maternal CMV viremia was not associated with infant hospitalization and/or death by 24 months. CONCLUSIONS Approximately 1 in 6 HIV-positive women in Botswana had detectable CMV DNA in blood at delivery. The presence of maternal CMV viremia had a borderline association with adverse birth outcomes but not with 24-month morbidity or mortality in HIV-exposed uninfected children.
Collapse
Affiliation(s)
- Natasha O Moraka
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Sikhulile Moyo
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Gloria Mayondi
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | | | - Maryanne Ibrahim
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Christiana Smith
- Pediatric Infectious Diseases University of Colorado Denver, Denver, CO
| | - Adriana Weinberg
- Pediatric Infectious Diseases University of Colorado Denver, Denver, CO
| | - Shaobing Li
- Pediatric Infectious Diseases University of Colorado Denver, Denver, CO
| | - Prisca K Thami
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Betsy Kammerer
- Department of Psychiatry, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Gbolahan Ajibola
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Rosemary Musonda
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Roger Shapiro
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Harvard Medical School, Boston, MA
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, MA
| | - Simani Gaseitsiwe
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Shahin Lockman
- Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Harvard Medical School, Boston, MA
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
44
|
Fry SHL, Barnabas SL, Cotton MF. Tuberculosis and HIV-An Update on the "Cursed Duet" in Children. Front Pediatr 2019; 7:159. [PMID: 32211351 PMCID: PMC7073470 DOI: 10.3389/fped.2019.00159] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/04/2019] [Indexed: 11/13/2022] Open
Abstract
HIV and tuberculosis (TB) often occur together with each exacerbating the other. Improvements in vertical transmission prevention has reduced the number of HIV-infected children being born and early antiretroviral therapy (ART) protects against tuberculosis. However, with delayed HIV diagnosis, HIV-infected infants often present with tuberculosis co-infection. The number of HIV exposed uninfected children has increased and these infants have high exposure to TB and may be more immunologically vulnerable due to HIV exposure in utero. Bacillus Calmette-Guérin (BCG) immunization shortly after birth is essential for preventing severe TB in infancy. With early infant HIV diagnosis and ART, disseminated BCG is no longer an issue. TB prevention therapy should be implemented for contacts of a source case and for all HIV-infected individuals over a year of age. Although infection can be identified through skin tests or interferon gamma release assays, the non-availability of these tests should not preclude prevention therapy, once active TB has been excluded. Therapeutic options have moved from isoniazid only for 6-9 months to shorter regimens. Prevention therapy after exposure to a source case with resistant TB should also be implemented, but should not prevent pivotal prevention trials already under way. A microbiological diagnosis for TB remains the gold standard because of increasing drug resistance. Antiretroviral therapy for rifampicin co-treatment requires adaptation for those on lopinavir-ritonavir, which requires super-boosting with additional ritonavir. For those with drug resistant TB, the main problems are identification and overlapping toxicity between antiretroviral and anti-TB therapy. In spite of renewed focus and improved interventions, infants are still vulnerable to TB.
Collapse
Affiliation(s)
| | | | - Mark F. Cotton
- Family Centre for Research with Ubuntu (FAM-CRU), Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
45
|
le Roux DM, Nicol MP, Myer L, Vanker A, Stadler JAM, von Delft E, Zar HJ. Lower Respiratory Tract Infections in Children in a Well-vaccinated South African Birth Cohort: Spectrum of Disease and Risk Factors. Clin Infect Dis 2019; 69:1588-1596. [DOI: 10.1093/cid/ciz017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 03/04/2019] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Childhood lower respiratory tract infections (LRTIs) cause substantial morbidity and under-5 child mortality. The epidemiology of LRTI is changing in low- and middle-income countries with expanding access to conjugate vaccines, yet there are few data on the incidence and risk factors for LRTI in these settings.
Methods
A prospective birth cohort enrolled mother–infant pairs in 2 communities near Cape Town, South Africa. Active surveillance for LRTI was performed for the first 2 years of life over 4 respiratory seasons. Comprehensive data collection of risk factors was done through 2 years of life. World Health Organization definitions were used to classify clinical LRTI and chest radiographs.
Results
From March 2012 to February 2017, 1143 children were enrolled and followed until 2 years of age. Thirty-two percent of children were exposed to antenatal maternal smoking; 15% were born at low birth weights. Seven hundred ninety-five LRTI events occurred in 429 children by February 2017; incidence of LRTI was 0.51 and 0.25 episodes per child-year in the first and second years of life, respectively. Human immunodeficiency virus (HIV)–exposed, uninfected infants (vs HIV-unexposed infants) were at increased risk of hospitalized LRTI in the first 6 months of life. In regression models, male sex, low birth weight, and maternal smoking were independent risk factors for both ambulatory and hospitalized LRTI; delayed or incomplete vaccination was associated with hospitalized LRTI.
Conclusions
LRTI incidence was high in the first year of life, with substantial morbidity. Strategies to ameliorate harmful exposures are needed to reduce LRTI burden in vulnerable populations.
Collapse
Affiliation(s)
- David M le Roux
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital and Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
- Department of Paediatrics, New Somerset Hospital, Cape Town, South Africa
| | - Mark P Nicol
- Division of Medical Microbiology, University of Cape Town and National Health Laboratory Service, Cape Town, South Africa
| | - Landon Myer
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Aneesa Vanker
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital and Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Jacob A M Stadler
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital and Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Eckart von Delft
- Department of Paediatrics, Paarl Hospital, Cape Town, South Africa
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital and Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
46
|
Huang J, Eskenazi B, Bornman R, Rauch S, Chevrier J. Maternal Peripartum Serum DDT/E and Urinary Pyrethroid Metabolite Concentrations and Child Infections at 2 Years in the VHEMBE Birth Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:067006. [PMID: 29906263 PMCID: PMC6108579 DOI: 10.1289/ehp2657] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 05/22/2023]
Abstract
BACKGROUND Indoor residual spraying (IRS) of insecticides, conducted in low- and middle-income countries to control malaria, may result in high exposure to dichlorodiphenyltrichloroethane (DDT), its breakdown product dichlorodiphenyldichloroethylene (DDE), or pyrethroids. Animal studies suggest in utero exposure to these chemicals may increase childhood infection frequency. OBJECTIVES We investigated associations between maternal DDT/E and pyrethroid metabolite concentration and child infection associations in an IRS setting in which susceptibility factors are common and infections are leading causes of child morbidity and mortality. METHODS Using gas chromatography-mass spectrometry, we measured serum DDT/E and urinary pyrethroid metabolite concentrations in peripartum samples from 674 women participating in the Venda Health Examination of Mother, Babies and their Environment (VHEMBE) study. Counts of persistent child fevers, otitis media, and severe sore throat between 1 and 2 y of age were ascertained from maternal interviews. Associations between DDT/E and pyrethroid metabolite concentrations and infections were estimated using zero-inflated Poisson regression. We estimated relative excess risks due to interaction (RERI) with poverty, maternal energy intake, and maternal HIV status. RESULTS Concentrations of DDT/E, particularly p,p'-DDE, were associated with higher rates of persistent fevers [IRR=1.21 (95% CI: 1.01, 1.46)], for a 10-fold increase in p,p'-DDE). This association was stronger among children from households below versus above the South African food poverty line [IRR=1.31 (95% CI: 1.08, 1.59) vs. IRR=0.93 (95% CI: 0.69, 1.25), respectively] and for children whose mothers had insufficient versus sufficient caloric intake during pregnancy [IRR=1.30 (95% CI: 1.07, 1.58) vs. IRR=0.96 (95% CI: 0.72, 1.28), respectively]. CONCLUSIONS In utero IRS insecticide exposure may increase childhood infection rates. This was particularly apparent among children from poorer households or whose mothers had low energy intake during pregnancy. https://doi.org/10.1289/EHP2657.
Collapse
Affiliation(s)
- Jonathan Huang
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Brenda Eskenazi
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California at Berkeley, Berkeley, California, USA
| | - Riana Bornman
- Centre for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Stephen Rauch
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California at Berkeley, Berkeley, California, USA
| | - Jonathan Chevrier
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
47
|
Wang YH, Shen XD. Human immunodeficiency virus infection and mortality risk among lung cancer patients: A systematic review and meta-analysis. Medicine (Baltimore) 2018; 97:e0361. [PMID: 29642182 PMCID: PMC5908612 DOI: 10.1097/md.0000000000010361] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Previous studies have suggested that patients with human immunodeficiency virus (HIV) infection are at higher risk of lung cancer, but the impact of HIV infection on the risk of mortality among lung cancer patients is still unclear. We conducted a systematic review and meta-analysis to clarify the association between HIV infection and mortality risk among lung cancer patients. METHODS PubMed and Embase databases were searched to identify studies assessing the association between HIV infection and mortality risk among lung cancer patients. Only studies reporting adjusted relative risk (RR) of mortality among lung cancer patients with HIV infection were included. Meta-analysis of random-effect model was utilized to calculate the pooled RR with 95% confidence interval (CI). RESULTS Twelve cohort studies were finally included. Compared with lung cancer patients without HIV infection, the pooled RR of mortality among lung cancer patients with HIV infection was 1.48 (95% CI, 1.22-1.78, P < .001; I = 88.6%). After excluding 2 studies with low quality, HIV infection was still significantly associated with an elevated risk of mortality among lung cancer patients (RR = 1.51, 95% CI, 1.25-1.82, P < .001; I = 89.8%). Sensitivity analysis showed that the statistical significance of the pooled RR was not changed by excluding any one study. CONCLUSION The outcomes from the meta-analysis provide strong evidence for the elevated risk of mortality among lung cancer patients with HIV infection, and HIV infection is an important prognostic factor in lung cancer patients.
Collapse
Affiliation(s)
- Yue-Hua Wang
- Department of Respiration, Jinhua People's Hospital, Jinhua
| | - Xiang-Di Shen
- Faculty of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
48
|
Baroncelli S, Galluzzo CM, Liotta G, Andreotti M, Mancinelli S, Mphwere R, Bokola E, Amici R, Marazzi MC, Palombi L, Lucaroni F, Giuliano M. Deficit of IgG2 in HIV-positive pregnant women is responsible of inadequate IgG2 levels in their HIV-uninfected children in Malawi. Med Microbiol Immunol 2018; 207:175-182. [PMID: 29488063 DOI: 10.1007/s00430-018-0537-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/17/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Transplacental passage of IgGs is impaired in HIV + pregnant women, possibly determining an inadequate immunological protection in their children. We aimed to determine the impact of maternal immunological IgG profile and immunoactivation status on the efficiency of transplacental passage of IgG subclasses in HIV + mothers. METHODS 16 mother/infants pairs were studied in Malawi. Mothers received antiretroviral therapy (ART) from the third trimester of pregnancy. Determinations of pre-ART levels of maternal sCD14, of IgG subclasses in mothers at delivery and in their 1-month-old infants, were performed using commercial ELISA kits. RESULTS At delivery, after a median of 10 weeks of ART, 12/16 mothers were hypergammaglobulinemic, with IgG levels (20.5 mg/ml, 95% CI:18.8-26.8) directly correlated to the plasmatic levels of sCD14 (r = 0.640, p = 0.014). IgG1 levels (17.9 mg/ml) accounted for 82% of IgG, IgG3 and IgG4 levels were in the normal range. A profound deficit of IgG2 was observed both in mothers (0.60 mg/ml) and in infants (0.14 mg/ml). Placental transfer ratio (range 0.16-0.42) did not show a selective impairment between the different IgG subclasses. The transplacental passage of all IgG subclasses was decreased in the presence of maternal IgG over 16 mg/ml (significantly for IgG1, p = 0.031) and of high levels of sCD14 (p = 0.063). CONCLUSIONS Transplacental passage was reduced for all IgG subclasses and inversely correlated to high levels of maternal IgGs and to the degree of immunoactivation. The profound depression of IgG2 in mothers suggests that IgG2 neonatal levels mostly reflect the maternal deficit rather than a selective impairment of IgG2 transfer.
Collapse
Affiliation(s)
- Silvia Baroncelli
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy.
| | - Clementina Maria Galluzzo
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| | - Giuseppe Liotta
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier, 1 00133, Rome, Italy
| | - Mauro Andreotti
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| | - Sandro Mancinelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier, 1 00133, Rome, Italy
| | - Robert Mphwere
- DREAM Program, Community of S. Egidio, P.O. Box 30355, Blantyre, Malawi
| | - Enok Bokola
- DREAM Program, Community of S. Egidio, P.O. Box 30355, Blantyre, Malawi
| | - Roberta Amici
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| | | | - Leonardo Palombi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier, 1 00133, Rome, Italy
| | - Francesca Lucaroni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier, 1 00133, Rome, Italy
| | - Marina Giuliano
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| |
Collapse
|
49
|
Ajaykumar A, Soudeyns H, Kakkar F, Brophy J, Bitnun A, Alimenti A, Albert AYK, Money DM, Côté HCF. Leukocyte Telomere Length at Birth and During the Early Life of Children Exposed to but Uninfected With HIV After In Utero Exposure to Antiretrovirals. J Infect Dis 2018; 217:710-720. [PMID: 29228317 PMCID: PMC5853286 DOI: 10.1093/infdis/jix618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022] Open
Abstract
Background Maternal combination antiretroviral therapy (cART) during pregnancy could impact the health of human immunodeficiency virus (HIV)-exposed, HIV-uninfected (HEU) children, because some antiretrovirals cross the placenta and can inhibit telomerase. Our objective was to compare leukocyte telomere length (LTL) in HEU children and HIV-unexposed, HIV-uninfected (HUU) children at birth and in early life and to investigate any relationship with cART exposure. Methods HEU and HUU children's blood LTL was compared cross-sectionally at birth, and during the first three years of life. Longitudinal HEU LTL dynamics was evaluated over that same period. Results At birth, the LTL in HEU children (n = 114) was not shorter than that in HUU children (n = 86), but female infants had longer LTL than male infants. Maternal cART (duration or type) showed no association with shorter infant LTL. Among 214 HEU children age- and sex-matched at a 1:1 ratio to HUU children, LTL declined similarly in both groups. In a longitudinal analysis, LTL attrition in HEU children was rapid from birth to 1 year of age and gradual thereafter. Zidovudine prophylaxis did not significantly alter LTL. Conclusions Our results indicate that from birth to 3 years of age, the LTL in HEU children is not negatively affected by exposure to maternal HIV infection and cART, at least not to the regimens used within this Canadian cohort, a reassuring finding.
Collapse
Affiliation(s)
- Abhinav Ajaykumar
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, Canada
| | - Hugo Soudeyns
- Unité d’immunopathologie virale, Centre de Recherche du CHU Sainte-Justine, Montreal, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Fatima Kakkar
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Canada
| | - Jason Brophy
- Department of Pediatrics, Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, Canada
| | - Ari Bitnun
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Ariane Alimenti
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Women’s Hospital and Health Centre, Vancouver, Canada
| | - Arianne Y K Albert
- BC Women’s Hospital and Health Centre, Vancouver, Canada
- Women’s Health Research Institute, Vancouver, Canada
| | - Deborah M Money
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
- BC Women’s Hospital and Health Centre, Vancouver, Canada
- Women’s Health Research Institute, Vancouver, Canada
| | - Hélène C F Côté
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, Canada
- Women’s Health Research Institute, Vancouver, Canada
| |
Collapse
|
50
|
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
|