1
|
Azevedo JF, Joyner G, Kundu S, Samanta K, Gomes-Solecki M. Maternal transfer of oral vaccine induced anti-OspA antibodies protects Peromyscus spp. from tick-transmitted Borrelia burgdorferi. Infect Immun 2025:e0021625. [PMID: 40387454 DOI: 10.1128/iai.00216-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
The efficacy and duration of passive immunity protection depend on maternal antibody levels and transfer efficiency. We investigated whether oral vaccination of Peromyscus leucopus dams with recombinant outer surface protein A (OspA)-expressing Escherichia coli could induce maternal transfer of anti-OspA antibodies and protect pups from Borrelia burgdorferi challenge. Dams were vaccinated until breeding pairs were created (i), until parturition (ii), and until pups were 2 weeks old (iii). Pups were challenged with nymphal Ixodes scapularis-transmitted B. burgdorferi at ~4 weeks of age. Anti-OspA IgG was quantified in dams and pups, and anti-B. burgdorferi IgG was quantified in pups. B. burgdorferi burden was assessed by flaB quantitative PCR in pups' tissues ~4 weeks after tick challenge, and viability of B. burgdorferi was assessed by culture of the heart tissue. P. leucopus pups born to dams vaccinated until breeding had low serologic anti-OspA antibody and were not protected from tick-transmitted B. burgdorferi infection. However, when dams' vaccination extended until parturition and until pups were 2 weeks old, significant anti-OspA antibody transfer and protection from B. burgdorferi infection occurred. This was evidenced by the absence of antibody to B. burgdorferi PepVF, absence of B. burgdorferi flaB DNA in heart and bladder tissues, and absence of flaB in culture from heart tissues from pups euthanized >9 weeks after birth. We show that the transfer of anti-OspA antibodies from vaccinated P. leucopus dams to offspring prevents tick transmission and infection dynamics of B. burgdorferi in the major reservoir host of this spirochete in the USA.IMPORTANCEThis study contributes to our understanding of how interventions based in reservoir-targeted outer surface protein A vaccines designed to block transmission of B. burgdorferi from infected Ixodes scapularis ticks may disrupt the enzootic cycle of this spirochete and reduce incidence of Lyme disease.
Collapse
Affiliation(s)
- Jose F Azevedo
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Greg Joyner
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Suman Kundu
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Kamalika Samanta
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Maria Gomes-Solecki
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
2
|
Bahadir Z, Narayan P, Wolters R, Permar SR, Fouda G, Hessell AJ, Haigwood NL. Monoclonal Antibodies for Pediatric Viral Disease Prevention and Treatment. Pediatrics 2025; 155:e2024068690. [PMID: 40174915 DOI: 10.1542/peds.2024-068690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 02/27/2025] [Indexed: 04/04/2025] Open
Abstract
Medical advancements over the last century have improved our ability to treat pediatric infectious diseases, significantly reducing associated morbidity and mortality worldwide. Although vaccines have been pivotal in this progress, many viral pathogens still do not currently have effective vaccines. The COVID-19 pandemic highlighted the need for rapid responses to emerging viral pathogens and introduced new tools to combat them. This review addresses human monoclonal antibodies (mAbs) as a strategy for treating and preventing viral infections in pediatric populations. We discuss previously used and currently available mAbs and advancements in mAb discovery. We address the future of mAb therapy by describing novel approaches in drug production and delivery platforms in addition to alternative antibody classes. Finally, we review the challenges and limitations of mAb therapy development for newborns and children.
Collapse
Affiliation(s)
- Zeynep Bahadir
- Department of Pediatrics, Weill Cornell Medicine, Cornell University, New York, New York
| | - Priyanka Narayan
- Department of Pediatrics, Weill Cornell Medicine, Cornell University, New York, New York
| | - Rachael Wolters
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, Cornell University, New York, New York
| | - Genevieve Fouda
- Department of Pediatrics, Weill Cornell Medicine, Cornell University, New York, New York
| | - Ann J Hessell
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Nancy L Haigwood
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| |
Collapse
|
3
|
Azevedo JF, Joyner G, Kundu S, Samanta K, Gomes-Solecki M. Maternal Transfer of Oral Vaccine Induced Anti-OspA Antibodies Protects Peromyscus spp Pups from Tick-Transmitted Borrelia burgdorferi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639966. [PMID: 40060546 PMCID: PMC11888318 DOI: 10.1101/2025.02.24.639966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
The efficacy and duration of passive immunity protection depends on maternal antibody levels and transfer efficiency. We investigated whether oral vaccination of Peromyscus leucopus dams with recombinant OspA-expressing E. coli could induce maternal transfer of anti-OspA antibodies and protect pups from Borrelia burgdorferi challenge. Dams were vaccinated until breeding pairs were created (i), until parturition (ii), and until pups were 2 weeks old (iii). Pups were challenged with nymphal I. scapularis -transmitted B. burgdorferi at ∼ 4 weeks of age. Anti-OspA IgG were quantified in dams and pups, and anti -B. burgdorferi IgG were quantified in pups. B. burgdorferi burden was assessed by flaB qPCR in pups' tissues ∼ 4 weeks after tick challenge and viability of B. burgdorferi was assessed by culture of heart tissue. P. leucopus pups born to dams vaccinated until breeding had low serologic anti-OspA antibody and were not protected from tick transmitted B. burgdorferi infection. However, when dams' vaccination extended until parturition and until pups were two weeks old, significant anti-OspA antibody transfer and protection from B. burgdorferi infection occurred. This was evidenced by absence of antibody to B. burgdorferi PepVF, absence of B. burgdorferi flaB DNA in heart and bladder tissues, and absence of flaB in culture from heart tissues from pups euthanized >9 weeks after birth. We show that transfer of anti-OspA antibodies from vaccinated P. leucopus dams to offspring prevents tick transmission and infection dynamics of B. burgdorferi in the major reservoir host of this spirochete in the United States. Importance This study contributes to our understanding of how interventions based in reservoir targeted OspA vaccines designed to block transmission of B. burgdorferi from infected I. scapularis ticks may disrupt the enzootic cycle of this spirochete and reduce incidence of Lyme disease.
Collapse
|
4
|
Maltezou PG, Kourkouni E, Kousi D, Hadjichristodoulou C, Dadouli A, Briana D, Papaevangelou V. Knowledge, Attitudes, and Practices Regarding Influenza and Pertussis Immunization During Pregnancy in Greece. Vaccines (Basel) 2025; 13:347. [PMID: 40333220 PMCID: PMC12030942 DOI: 10.3390/vaccines13040347] [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: 01/30/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND/OBJECTIVES Vaccination against influenza and pertussis in pregnant women protects the mother and child through the transfer of protective antibodies across the placenta. However, pregnant women's vaccine hesitancy is a major barrier to achieve satisfactory vaccination coverage in many developed countries. METHODS Greek pregnant women's vaccination knowledge, attitudes, and practices were recorded. Structured questionnaires were administered to mothers of infants under the age of 12 months through their pediatricians. Sampling across the country's districts was applied to achieve geographic representativeness. RESULTS Questionnaires from 474 mothers were collected. Their mean age was 34 (±5) years. Vaccination uptake was 16.8% and 45.7%, for pertussis and influenza, respectively. During their recent pregnancy, 68.9% and 27.1% of the responders had been informed by their gynecologists regarding influenza and pertussis maternal immunization, respectively, indicating that gynecologists miss out on informing a significant rate of pregnant women. According to multiple logistic regression, women who gave birth during spring (OR: 2.29 vs. winter delivery, p = 0.042) and those with an MSc or PhD (OR: 2.93 vs. school graduates, p = 0.015) were more likely to receive influenza vaccination. Factors favoring influenza vaccination included doctor's recommendation (OR: 18.86, p < 0.001), being not/somewhat afraid of potential vaccine side effects during pregnancy (OR: 2.09, p = 0.012), considering the flu as relatively/very dangerous during pregnancy (OR: 8.05, p < 0.001), and considering the flu vaccine as relatively/completely safe (OR: 4.37, p < 0.001). Doctor's recommendation (OR: 29.55, p < 0.001) and considering pertussis a relatively/very serious risk to the mother's health during pregnancy (OR: 6.00, p = 0.002) were factors associated with pertussis vaccination during pregnancy. CONCLUSIONS The education of both expectant mothers and obstetricians is urgently needed in order to increase immunization coverage during pregnancy. The low influenza vaccination coverage among women delivering during winter and low pertussis immunization rates, in combination with low recommendation rates for both vaccines, strongly indicate that Greek obstetricians focus on maternal health alone. Their perspectives play an instrumental role in vaccine acceptance during pregnancy, shaping the immunization inclusion maps.
Collapse
Affiliation(s)
| | - Eleni Kourkouni
- Collaborative Center for Clinical Epidemiology and Outcomes Research (CLEO), 154 51 Athens, Greece
| | - Dimitra Kousi
- Collaborative Center for Clinical Epidemiology and Outcomes Research (CLEO), 154 51 Athens, Greece
| | | | - Aikaterini Dadouli
- Laboratory of Hygiene and Epidemiology, University of Thessaly, 382 21 Larissa, Greece
| | - Despoina Briana
- Third Department of Pediatrics, University Hospital Attikon, 124 62 Athens, Greece (V.P.)
| | | |
Collapse
|
5
|
Medoro AK, Puopolo KM. Transplacental Antibodies: Role of Maternal Vaccines and Immunity. Clin Perinatol 2025; 52:101-113. [PMID: 39892946 DOI: 10.1016/j.clp.2024.10.007] [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] [Indexed: 02/04/2025]
Abstract
Transplacental transfer of maternal antibody in utero is a key component of neonatal immunity. This transfer of maternal antibody across the placenta provides temporary protection from bacterial and viral pathogens that threaten newborn health. Maternal vaccination before and during pregnancy may be used to boost maternal immunity and optimize neonatal protection from infection and serious illness.
Collapse
Affiliation(s)
- Alexandra K Medoro
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Infectious Disease, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Karen M Puopolo
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Section on Newborn Medicine, Pennsylvania Hospital, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Baroncelli S, Galluzzo CM, Orlando S, Luhanga R, Mphwere R, Kavalo T, Amici R, Floridia M, Andreotti M, Ciccacci F, Marazzi MC, Giuliano M. Low levels of pertussis- and measles-specific IgG antibodies in 6-week-old HIV-exposed and -unexposed Malawian infants: implications for vaccination strategies and role of long term HIV therapy. J Trop Pediatr 2025; 71:fmaf013. [PMID: 40064194 DOI: 10.1093/tropej/fmaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Serological studies in infants can provide valuable information on the degree of protection conferred by IgG maternal passive transfer during early life. If infant levels are inadequate, protection may be incomplete, increasing the risk of life-threatening diseases such as pertussis and measles, before immunization completion. In addition, HIV infection, -highly prevalent in African countries like Malawi-may impair transplacental antibody transfer. We determined anti-Pertussis Toxin (PT) and anti-measles IgG in 86 6-week-old infants, born to mothers living with HIV (HIV-exposed uninfected, HEU, n = 58) and to HIV-negative mothers (HIV-unexposed uninfected, HUU, n = 28). The HEU group was divided into two subgroups: Infants born to mothers who initiated antiretroviral therapy (ART) during pregnancy (Short-ART, SA-HEU group, n = 29) or already in stable ART (Long-term ART, LA-HEU group, n = 29). The mean anti-PT and anti-measles IgG levels (1.97 IU/ml and 32.9 mIU/ml, respectively) were comparable between the HUU and HEU infants. Overall, only 12.8% and 18.6% of all infants had IgG levels above the protective thresholds for pertussis and measles, respectively. The duration of ART significantly influenced the infant's serological profile, with SA-HEU infants showing significantly lower IgG levels compared to both HUU and LA-HEU infants. Protecting infants during early life remains a significant health challenge in many middle and low-income countries. Achieving better early serological protection requires the implementation of diverse vaccination strategies. This study emphasizes the crucial importance for women living with HIV to be on stable ART before pregnancy.
Collapse
Affiliation(s)
- Silvia Baroncelli
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome Italy
| | | | - Stefano Orlando
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Richard Luhanga
- DREAM Program, Community of S. Egidio, 312224, Blantyre, Malawi
| | - Robert Mphwere
- DREAM Program, Community of S. Egidio, 312224, Blantyre, Malawi
| | - Thom Kavalo
- DREAM Program, Community of S. Egidio, 312224, Blantyre, Malawi
| | - Roberta Amici
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome Italy
| | - Marco Floridia
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome Italy
| | - Mauro Andreotti
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome Italy
| | - Fausto Ciccacci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy
| | | | - Marina Giuliano
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome Italy
| |
Collapse
|
7
|
Brown JA, Bashir H, Zeng MY. Lifelong partners: Gut microbiota-immune cell interactions from infancy to old age. Mucosal Immunol 2025:S1933-0219(25)00006-6. [PMID: 39862964 DOI: 10.1016/j.mucimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Our immune system and gut microbiota are intricately coupled from birth, both going through maturation during early life and senescence during aging almost in a synchronized fashion. The symbiotic relationship between the human host and microbiota is critically dependent on a healthy immune system to keep our microbiota in check, while the microbiota provides essential functions to promote the development and fitness of our immune system. The partnership between our immune system and microbiota is particularly important during early life, when microbial ligands and metabolites shape the development of the immune cells and immune tolerance; during aging, having sufficient beneficial gut bacteria is critical for the maintenance of intact mucosal barriers, immune metabolic fitness, and strong immunity against pathogens. The immune system during childhood is programmed, with the support of the microbiota, to develop robust immune tolerance, and limit autoimmunity and metabolic dysregulation, which are prevalent during aging. This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age, with the goal to gain a better understanding of potential strategies to leverage the gut microbiota to combat age-related immune decline.
Collapse
Affiliation(s)
- Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Hilal Bashir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, United States.
| |
Collapse
|
8
|
Ogasawara R, Urashima M. Cord blood IgG for respiratory syncytial virus and subsequent infection during the COVID-19 pandemic. Medicine (Baltimore) 2025; 104:e41110. [PMID: 40184109 PMCID: PMC11709157 DOI: 10.1097/md.0000000000041110] [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: 07/11/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 04/05/2025] Open
Abstract
The resurgence of respiratory syncytial virus (RSV) infection among Japanese infants during the coronavirus disease 2019 pandemic might be due to a decrease in cord blood anti-RSV immunoglobulin G (IgG) positivity resulting from reduced maternal RSV exposure. This study examined changes in the positivity before and during the pandemic to clarify the relationship between this positivity and infantile/severe RSV infections. Data from a prospective cohort study conducted in Tokyo, Japan, involving mother-child pairs of infants born between February 2019 and August 2022 were reanalyzed. Cord blood anti-RSV IgG levels measured by enzyme-linked immunosorbent assay were classified as positive, gray zone, and negative. We examined the relationship between antibody positivity and infantile (≤12 months old) and/or severe RSV infections as diagnosed by pediatricians. A total of 319 families participated. There was a significant decrease in cord blood anti-RSV IgG positivity from 65.1% in 2019 to 50.9% in 2022 (P = .02). Among infants followed up until their 2nd birthday, 43 (33.1%) were diagnosed with RSV infections. Of these cases, 5 were infantile/severe and occurred among the 50 infants classified as negative or gray zone, while none were observed among the 80 infants classified as positive (P = .004). Infantile/severe RSV infection was identified in 4 (16.7%) born during the pandemic, representing a significantly higher rate than 1 (0.9%) born before the pandemic (P < .001). This study revealed an association between lower cord blood anti-RSV IgG positivity during the coronavirus disease 2019 pandemic compared to before the pandemic, and an increased risk of subsequent infant/severe RSV infections.
Collapse
Affiliation(s)
- Ritsuko Ogasawara
- Division of Molecular Epidemiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Mitsuyoshi Urashima
- Division of Molecular Epidemiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Rizzuto G. B Cell Responses to the Placenta and Fetus. ANNUAL REVIEW OF PATHOLOGY 2025; 20:33-58. [PMID: 39264989 PMCID: PMC11912550 DOI: 10.1146/annurev-pathmechdis-111523-023459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Pregnancy has fascinated immunologists ever since Peter Medawar's observation that reproduction runs contrary to the founding tenets of immunology. During healthy pregnancy, maternal B cells interact with antigens of the foreign conceptus (placenta and fetus) yet do not elicit rejection. Instead, robust and redundant fetomaternal tolerance pathways generally prevent maternal B cells and antibodies from harming the placenta and fetus. Fetomaternal tolerance is not absolute, and unfortunately there exist several pregnancy complications that arise from breaks therein. Here, important historic and recent developments in the field of fetomaternal tolerance pertaining to maternal B cells and antibodies are reviewed. General rules from which to conceptualize humoral tolerance to the placenta and fetus are proposed. Significant but underexplored ideas are highlighted and topics for future research are suggested, findings from which are predicted to provide insight into the fundamental nature of tolerance and bolster efforts to combat immune-mediated pregnancy complications.
Collapse
Affiliation(s)
- Gabrielle Rizzuto
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA
- Department of Anatomic Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immuno-Oncology, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA;
| |
Collapse
|
10
|
Makan-Murphy N, Madhi SA, Dangor Z. Safety, Efficacy, and Effectiveness of Maternal Vaccination against Respiratory Infections in Young Infants. Semin Respir Crit Care Med 2024. [PMID: 39708836 DOI: 10.1055/a-2471-6906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2024]
Abstract
Lower respiratory tract infection (LRTI) is a major cause of neonatal morbidity and mortality worldwide. Maternal vaccination is an effective strategy in protecting young infants from LRTI, particularly in the first few months after birth when infant is most vulnerable, and most primary childhood vaccinations have not been administered. Additionally, maternal vaccination protects the mother from illness during pregnancy and the postnatal period, and the developing fetus from adverse outcomes such as stillbirth and prematurity. In this paper, we review the safety, efficacy, and effectiveness of maternal vaccines against LRTIs, such as pertussis, influenza, coronavirus disease 2019, and respiratory syncytial virus.
Collapse
Affiliation(s)
- Nisha Makan-Murphy
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ziyaad Dangor
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
11
|
Suominen H, Syrjänen K, Waterboer T, Grénman S, Syrjänen S, Louvanto K. Serum Immunoglobulin G Antibodies to Human Papillomavirus Type 6 L1, E2, E4, E6, and E7 Proteins Among Children Prospectively Followed up for 3 Years. J Infect Dis 2024; 230:e1207-e1213. [PMID: 38820118 PMCID: PMC11646598 DOI: 10.1093/infdis/jiae293] [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: 02/02/2024] [Revised: 04/20/2024] [Accepted: 05/30/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Current knowledge implicates that human papillomavirus (HPV) infection can be acquired at an early age. However, the role of HPV-specific passive immunization from mother to neonate is nearly unexplored, especially against the HPV early proteins. We analyzed immunoglobulin G (IgG) antibodies against HPV-6 early (E2, E4, E6, E7) and late (L1) proteins in children prospectively followed up for 3 years. METHODS A total of 272 children and their mothers from the Finnish Family HPV Study were included in these analyses. Serum samples were obtained from pregnant mothers at their third trimester and from newborn/infants at 1-, 2-, 6-, 12-, 24-, and 36-month visits after birth. Antibodies were analyzed by multiplex serology based on glutathione S-transferase fusion protein capture to fluorescent beads. RESULTS Maternal antibodies to all tested HPV-6 proteins were transferred to neonates, concordance between maternal and neonates' antibody levels being highly significant (P < .001). Seropositivity of HPV-6 L1 in the neonates declined during the first 6 months of life, whereas changes in the E protein antibodies were less obvious. After the maternal antibodies had vanished, seroconversion to HPV-6 L1 at 12 months (median) and to the HPV-6 E proteins between 23 and 35 months was observed. CONCLUSIONS IgG antibodies against HPV-6 E and L proteins are transferred from mothers to their children. Seroconversion against HPV-6 L1, E2, E4, E6, and E7 does occur in early childhood, as a sign of acquired HPV-6 infection by vertical or horizontal transmission starting at 12 months of age.
Collapse
Affiliation(s)
- Helmi Suominen
- Department of Obstetrics and Gynecology, Tampere University, Tampere
| | | | - Tim Waterboer
- Division of Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Seija Grénman
- Department of Obstetrics and Gynecology, Turku University Hospital, Turku
- Department of Oral Pathology and Oral Radiology, University of Turku, Turku
| | - Stina Syrjänen
- Department of Obstetrics and Gynecology, Turku University Hospital, Turku
- Department of Oral Pathology and Oral Radiology, University of Turku, Turku
| | - Karolina Louvanto
- Department of Obstetrics and Gynecology, Tampere University, Tampere
- Department of Oral Pathology and Oral Radiology, University of Turku, Turku
- Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
12
|
Zhang SL, McGann CM, Duranova T, Strysko J, Steenhoff AP, Gezmu A, Nakstad B, Arscott-Mills T, Bayani O, Moorad B, Tlhako N, Richard-Greenblatt M, Hu W, Planet PJ, Coffin SE, Silverman MA. Maternal and neonatal IgG against Klebsiella pneumoniae are associated with lower risk of neonatal sepsis: A case-control study of hospitalized neonates in Botswana. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003350. [PMID: 39637243 PMCID: PMC11620667 DOI: 10.1371/journal.pgph.0003350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
Sepsis is the leading postnatal cause of neonatal mortality worldwide. Globally Klebsiella pneumoniae is the leading cause of sepsis in hospitalized neonates. This study reports the development and evaluation of an ELISA for anti-Klebsiella IgG using dried blood spot (DBS) samples and evaluates the association of anti-Klebsiella IgG (anti-Kleb IgG) antibodies in maternal and neonatal samples with the risk of neonatal sepsis. Neonates and their mothers were enrolled at 0-96 hours of life in the neonatal unit of a tertiary referral hospital in Gaborone, Botswana and followed until death or discharge to assess for episodes of blood culture-confirmed neonatal sepsis. Neonates with sepsis had significantly lower levels of Kleb-IgG compared to neonates who did not develop sepsis (Mann-Whitney U, p = 0.012). Similarly, samples from mothers of neonates who developed sepsis tended to have less Kleb-IgG compared to mothers of controls. The inverse correlation between Kleb-IgG levels and all-cause bacteremia suggests that maternal Kleb-IgG may be protective through cross-reactivity with common bacterial epitopes. These data support the continued use of immunoglobulin assays using DBS samples to explore the role of passive immunity on neonatal sepsis risk and reaffirm the critical need for research supporting the development of maternal vaccines for neonatal sepsis.
Collapse
Affiliation(s)
- Siqi Linsey Zhang
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Carolyn M. McGann
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tereza Duranova
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Jonathan Strysko
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Paediatric & Adolescent Health, Faculties of Medicine & Health Sciences, University of Botswana, Gaborone, Botswana
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Andrew P. Steenhoff
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - Alemayehu Gezmu
- Department of Paediatric & Adolescent Health, Faculties of Medicine & Health Sciences, University of Botswana, Gaborone, Botswana
| | - Britt Nakstad
- Department of Paediatric & Adolescent Health, Faculties of Medicine & Health Sciences, University of Botswana, Gaborone, Botswana
| | - Tonya Arscott-Mills
- Department of Paediatric & Adolescent Health, Faculties of Medicine & Health Sciences, University of Botswana, Gaborone, Botswana
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - One Bayani
- Department of Paediatric & Adolescent Health, Faculties of Medicine & Health Sciences, University of Botswana, Gaborone, Botswana
| | - Banno Moorad
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - Nametso Tlhako
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - Melissa Richard-Greenblatt
- Hospital for Sick Children, Toronto, Canada
- Department of Laboratory and Pathobiology, University of Toronto, Toronto, Canada
| | - Weiming Hu
- Division of Gastroenterology, Hepatology and Nutrition Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- CHOP Microbiome Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Paul J. Planet
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Susan E. Coffin
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael A. Silverman
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
13
|
Vijayan K. K. V, De Paris K. Nonhuman primate models of pediatric viral diseases. Front Cell Infect Microbiol 2024; 14:1493885. [PMID: 39691699 PMCID: PMC11649651 DOI: 10.3389/fcimb.2024.1493885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Infectious diseases are the leading cause of death in infants and children under 5 years of age. In utero exposure to viruses can lead to spontaneous abortion, preterm birth, congenital abnormalities or other developmental defects, often resulting in lifelong health sequalae. The underlying biological mechanisms are difficult to study in humans due to ethical concerns and limited sample access. Nonhuman primates (NHP) are closely related to humans, and pregnancy and immune ontogeny in infants are very similar to humans. Therefore, NHP are a highly relevant model for understanding fetal and postnatal virus-host interactions and to define immune mechanisms associated with increased morbidity and mortality in infants. We will discuss NHP models of viruses causing congenital infections, respiratory diseases in early life, and HIV. Cytomegalovirus (CMV) remains the most common cause of congenital defects worldwide. Measles is a vaccine-preventable disease, yet measles cases are resurging. Zika is an example of an emerging arbovirus with devastating consequences for the developing fetus and the surviving infant. Among the respiratory viruses, we will discuss influenza and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We will finish with HIV as an example of a lifelong infection without a cure or vaccine. The review will highlight (i) the impact of viral infections on fetal and infant immune development, (ii) how differences in infant and adult immune responses to infection alter disease outcome, and emphasize the invaluable contribution of pediatric NHP infection models to the design of effective treatment and prevention strategies, including vaccines, for human infants.
Collapse
Affiliation(s)
- Vidya Vijayan K. K.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Center for AIDS Research, University of North Carolina, Chapel Hill, NC, United States
- Children’s Research Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
14
|
Semmes EC, Nettere DR, Nelson AN, Hurst JH, Cain DW, Burt TD, Kurtzberg J, Reeves RK, Coyne CB, Fouda GG, Pollara J, Permar SR, Walsh KM. In utero human cytomegalovirus infection expands NK-like FcγRIII+CD8+ T cells that mediate Fc antibody functions. J Clin Invest 2024; 135:e181342. [PMID: 39531313 PMCID: PMC11684805 DOI: 10.1172/jci181342] [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: 03/25/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Human cytomegalovirus (HCMV) profoundly impacts host T and NK cells across the lifespan, yet how this common congenital infection modulates developing fetal immune cell compartments remains underexplored. Using cord blood from neonates with and without congenital HCMV (cCMV) infection, we identify an expansion of Fcγ receptor III-expressing (FcγRIII-expressing) CD8+ T cells following HCMV exposure in utero. Most FcγRIII+CD8+ T cells express the canonical αβ T cell receptor (TCR), but a proportion express noncanonical γδ TCR. FcγRIII+CD8+ T cells are highly differentiated and have increased expression of NK cell markers and cytolytic molecules. Transcriptional analysis reveals FcγRIII+CD8+ T cells upregulate T-bet and downregulate BCL11B, known transcription factors that govern T/NK cell fate. We show that FcγRIII+CD8+ T cells mediate antibody-dependent IFN-γ production and degranulation against IgG-opsonized target cells, similar to NK cell antibody-dependent cellular cytotoxicity (ADCC). FcγRIII+CD8+ T cell Fc effector functions were further enhanced by IL-15, as has been observed in neonatal NK cells. Our study reveals that FcγRIII+CD8+ T cells elicited in utero by HCMV infection can execute Fc-mediated effector functions bridging cellular and humoral immunity and may be a promising target for antibody-based therapeutics and vaccination in early life.
Collapse
Affiliation(s)
- Eleanor C. Semmes
- Boston Children’s Hospital/Boston Medical Center, Boston, Massachusetts, USA
- Medical Scientist Training Program, and
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Danielle R. Nettere
- Medical Scientist Training Program, and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ashley N. Nelson
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jillian H. Hurst
- Children’s Health and Discovery Initiative
- Division of Infectious Diseases, and
| | - Derek W. Cain
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Trevor D. Burt
- Children’s Health and Discovery Initiative
- Division of Neonatology, Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Children’s Health and Discovery Initiative
- Carolinas Cord Blood Bank, Marcus Center for Cellular Cures, Durham, North Carolina, USA
| | - R. Keith Reeves
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Human Systems Immunology, and
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Carolyn B. Coyne
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Children’s Health and Discovery Initiative
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Children’s Health and Discovery Initiative
- Division of Infectious Diseases, and
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Kyle M. Walsh
- Children’s Health and Discovery Initiative
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| |
Collapse
|
15
|
Charlesworth JEG. Mind the (preterm) gap: inequality in the UK's current RSV immunisation approach will leave many preterm babies unprotected against RSV this winter. Arch Dis Child 2024:archdischild-2024-327741. [PMID: 39379140 DOI: 10.1136/archdischild-2024-327741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024]
Affiliation(s)
- James E G Charlesworth
- Department of Paediatrics, Medical Sciences Division, University of Oxford, Oxford, UK
- Paediatrics, Milton Keynes University Hospital NHS Foundation Trust, Milton Keynes, UK
| |
Collapse
|
16
|
Lee HY, Nazmul T, Lan J, Oyoshi MK. Maternal influences on offspring food allergy. Immunol Rev 2024; 326:130-150. [PMID: 39275992 PMCID: PMC11867100 DOI: 10.1111/imr.13392] [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: 09/16/2024]
Abstract
The prevalence of allergies has been globally escalating. While allergies could appear at any age, they often develop in early life. However, the significant knowledge gap in the field is the mechanisms by which allergies affect certain people but not others. Investigating early factors and events in neonatal life that have a lasting impact on determining the susceptibilities of children to develop allergies is a significant area of the investigation as it promotes the understanding of neonatal immune system that mediates tolerance versus allergies. This review focuses on the research over the recent 10 years regarding the potential maternal factors that influence offspring allergies with a view to food allergy, a potentially life-threatening cause of anaphylaxis. The role of breast milk, maternal diet, maternal antibodies, and microbiota that have been suggested as key maternal factors regulating offspring allergies are discussed here. We also suggest future research area to expand our knowledge of maternal-offspring interactions on the pathogenesis of food allergy.
Collapse
Affiliation(s)
- Hwa Yeong Lee
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanuza Nazmul
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Jinggang Lan
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Michiko K. Oyoshi
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Jacob A, Shatila AO, Inshasi J, Massouh J, Mir R, Noori S, Yamout B. Disease modifying treatment guidelines for multiple sclerosis in the United Arab Emirates. Mult Scler Relat Disord 2024; 88:105703. [PMID: 38924933 DOI: 10.1016/j.msard.2024.105703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/13/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
The newly constituted National Multiple Sclerosis (MS) Society (NMSS)of the United Arab Emirates (UAE), set up a scientific committee to create a MS disease modifying treatment (DMT) guideline for UAE. The committee considered several unique features of the MS community in UAE including large number of expatriate population, wide variations in health insurance coverage, physician and patient preferences for DMT. The overall goal of the treatment guideline is to facilitate the most appropriate DMT to the widest number of patients. To this end it has adapted recommendations from various health systems and regulatory authorities into a pragmatic amalgamation of best practices from across the world. Importantly where data is unavailable or controversial, a common sense approach is taken rather than leave physicians and patients in limbo. The committee classifies MS into subcategories and suggests appropriate treatment choices. It recommends treatment of RIS and CIS with poor prognostic factors. It largely equates the efficacy and safety of DMT with similar mechanisms of action or drug classes e.g. ocrelizumab is similar to rituximab. It allows early switching of treatment for unambiguous disease activity and those with progression independent of relapses. Autologous hematopoietic stem cell transplantation can be offered to patients who fail one high efficacy DMT. Pragmatic guidance on switching and stopping DMT, DMT choices in pregnancy, lactation and pediatric MS have been included. It is expected that these guidelines will be updated periodically as new data becomes available.
Collapse
Affiliation(s)
- Anu Jacob
- Neurological Institute, Cleveland Clinic, Abu Dhabi, United Arab Emirates; The Walton Centre, Liverpool, United Kingdom.
| | - Ahmed Osman Shatila
- Department of Neurology, Sheikh Shakhbout Medical City Abu Dhabi, United Arab Emirates
| | - Jihad Inshasi
- Department of Neurology, Rashid Hospital and Dubai Medical College, Dubai Health Authority, Dubai, United Arab Emirates
| | - Joelle Massouh
- Neurology Institute and Multiple Sclerosis Centre, Harley Street Medical centre, Abu Dhabi, United Arab Emirates
| | - Ruquia Mir
- Abu Dhabi stem Cell Clinic, United Arab Emirates
| | - Suzan Noori
- University Hospital Sharjah, United Arab Emirates
| | - Bassem Yamout
- Neurology Institute and Multiple Sclerosis Centre, Harley Street Medical centre, Abu Dhabi, United Arab Emirates; American University of Beirut, Lebanon
| |
Collapse
|
18
|
Mata-Miranda MM, Martinez-Cuazitl A, Gutierrez-Cortes H, Cordero-Hernandez L, Guerrero-Ruiz M, Lopez-Reyes A, Rodriguez-Baez A, Vazquez-Zapien GJ. Analysis of the immune response using FTIR spectroscopy in mothers and their newborns with different vaccination schemes for COVID-19. Sci Rep 2024; 14:17308. [PMID: 39068230 PMCID: PMC11283522 DOI: 10.1038/s41598-024-68340-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
The SARS-CoV-2 outbreak has provoked more than 6 million deaths worldwide. The scarcity of effective treatments and its virulence converted the vaccines into an essential tool to face it. The most used vaccines were the mRNA, adenovirus vector, and inactivated whole-virus. However, nowadays, infants aged < 6 months are not eligible for any vaccines against COVID-19, and their immunization relies on passive immunity. In this research, we investigated the humoral and cellular immune response generated on newborns of SARS-CoV-2 vaccinated mothers with mRNA or viral vector (VV) vaccine employing Fourier transformed infrared (FTIR) spectroscopy in saliva samples. For this purpose, saliva samples of newborns and their mothers were collected; the population was divided into two groups, VV and mRNA, which were subdivided into three subgroups: before pregnancy (BP), at the first (FTP) and second (STP) trimesters of pregnancy. The samples were analyzed using FTIR spectroscopy, and the bands associated with the humoral and cellular immune responses, such as IgG, IgA, and IFN-γ were analyzed. The integrated areas were calculated and compared to elucidate the quantity of those immunoglobins and the cytokine. Likewise, the correlation of the humoral and cellular immune response between the newborns and their mothers and the correlation between cellular and humoral immune response was also evaluated. The VV vaccine produced a significant humoral and cellular immune response in newborns and their mothers when they received it at the STP compared with the mRNA vaccine, evidencing statistical significance. However, no correlation was observed between newborns and their mothers when the vaccine was applied in this trimester of pregnancy. When administered BP, the mRNA vaccine generated more humoral immunity in newborns and their mothers. Nevertheless, compared with the VV vaccine, it only showed statistical significance in the mothers, highlighting that IgG showed a moderate positive correlation between the newborns and their mothers.
Collapse
Affiliation(s)
- Monica M Mata-Miranda
- Escuela Militar de Medicina, Centro Militar de Ciencias de la Salud, Secretaría de la Defensa Nacional, 11200, Mexico City, Mexico.
| | - Adriana Martinez-Cuazitl
- Escuela Militar de Medicina, Centro Militar de Ciencias de la Salud, Secretaría de la Defensa Nacional, 11200, Mexico City, Mexico
| | - Hermes Gutierrez-Cortes
- Hospital Militar de Especialidades de la Mujer y Neonatología, Secretaría de la Defensa Nacional, 11200, Mexico City, Mexico
| | - Librado Cordero-Hernandez
- Hospital Militar de Especialidades de la Mujer y Neonatología, Secretaría de la Defensa Nacional, 11200, Mexico City, Mexico
| | - Melissa Guerrero-Ruiz
- Escuela Militar de Medicina, Centro Militar de Ciencias de la Salud, Secretaría de la Defensa Nacional, 11200, Mexico City, Mexico
- Escuela Superior de Medicina, Instituto Politécnico Nacional, 11340, Mexico City, Mexico
| | - Alberto Lopez-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, 14389, Mexico City, Mexico
| | - Alvaro Rodriguez-Baez
- Centro de Investigación y Desarrollo del Ejército y Fuerza Aérea Mexicanos, Secretaría de la Defensa Nacional, 11400, Mexico City, Mexico
| | - Gustavo J Vazquez-Zapien
- Escuela Militar de Medicina, Centro Militar de Ciencias de la Salud, Secretaría de la Defensa Nacional, 11200, Mexico City, Mexico.
- Centro de Investigación y Desarrollo del Ejército y Fuerza Aérea Mexicanos, Secretaría de la Defensa Nacional, 11400, Mexico City, Mexico.
| |
Collapse
|
19
|
Butler EA, Grandi SM, Matai L, Wang X, Cohen E, Ray JG. Differences in maternal-newborn ABO blood groups and risk of serious infant infection. QJM 2024; 117:512-519. [PMID: 38402542 PMCID: PMC11290255 DOI: 10.1093/qjmed/hcae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/11/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND During pregnancy, various maternal IgG antibodies are transferred to the developing fetus, some of which may protect the newborn against infection. If a mother and her fetus have different A, B or O (ABO) blood groups, then transferred maternal antibodies may plausibly protect the infant against infection. AIM To determine if maternal-newborn ABO blood group incongruence vs. congruence is associated with a lower risk of serious infection in the infant. DESIGN Retrospective population-based cohort. METHODS We used linked patient-level datasets for all singleton hospital livebirths from 2008 to 2022 in Ontario, Canada, with known maternal and newborn ABO blood groups. We used a dichotomous exposure state, either ABO blood group congruent (N = 114 507) or incongruent (N = 43 074). The main outcome of interest was the risk of serious infant infection within 27 days, and from 28 to 365 days, after birth. Cox proportional hazard models generated hazard ratios and 95% confidence intervals, and were adjusted for maternal age, world region of origin, residential income quintile and gestational age at birth. RESULTS Relative to maternal-newborn congruency, incongruent ABO blood group was associated with an adjusted hazard ratio of 0.88 (95% CI: 0.80-0.97) for serious neonatal infection within 27 days of birth, and 0.93 (95% CI: 0.90-0.96) for serious infection between 28 and 365 days after birth. CONCLUSIONS Maternal-newborn ABO incongruence may be associated with a lower relative risk of a serious infant infection within 27 days, and from 28 to 365 days, after birth.
Collapse
Affiliation(s)
- E A Butler
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
- ICES, Toronto, ON, Canada
| | - S M Grandi
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
- ICES, Toronto, ON, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | - X Wang
- ICES, Toronto, ON, Canada
| | - E Cohen
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
- ICES, Toronto, ON, Canada
| | - J G Ray
- ICES, Toronto, ON, Canada
- Department of Medicine, St Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
20
|
England RN, Drapeau EM, Alameh MG, Hosseinzadeh R, Weissman D, Hensley SE. Evaluation of mRNA-LNP and adjuvanted protein SARS-CoV-2 vaccines in a maternal antibody mouse model. NPJ Vaccines 2024; 9:110. [PMID: 38890316 PMCID: PMC11189435 DOI: 10.1038/s41541-024-00901-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Maternal antibodies (matAbs) protect against a myriad of pathogens early in life; however, these antibodies can also inhibit de novo immune responses against some vaccine platforms. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) matAbs are efficiently transferred during pregnancy and protect infants against subsequent SARS-CoV-2 infections. It is unknown if matAbs inhibit immune responses elicited by different types of SARS-CoV-2 vaccines. Here, we established a mouse model to determine if SARS-CoV-2 spike-specific matAbs inhibit immune responses elicited by recombinant protein and nucleoside-modified mRNA-lipid nanoparticle (mRNA-LNP) vaccines. We found that SARS-CoV-2 mRNA-LNP vaccines elicited robust de novo antibody responses in mouse pups in the presence of matAbs. Recombinant protein vaccines were also able to circumvent the inhibitory effects of matAbs when adjuvants were co-administered. While additional studies need to be completed in humans, our studies raise the possibility that mRNA-LNP-based and adjuvanted protein-based SARS-CoV-2 vaccines have the potential to be effective when delivered very early in life.
Collapse
Affiliation(s)
- Ross N England
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth M Drapeau
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Nsubuga R, Rujumba J, Nyende S, Kisaka S, Idro R, Nankunda J. Predictors of mortality among low birth weight neonates after hospital discharge in a low-resource setting: A case study in Uganda. PLoS One 2024; 19:e0303454. [PMID: 38861517 PMCID: PMC11166315 DOI: 10.1371/journal.pone.0303454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Most neonatal deaths occur among low birth weight infants. However, in resource-limited settings, these infants are commonly discharged early which further exposes them to mortality. Previous studies on morbidity and mortality among low birth weight infants after early discharge mainly focused on very low birth weight infants, and none described post-discharge neonatal mortality. This study aimed to determine the proportion and predictors of mortality among low birth weight neonates discharged from the Special Care Baby Unit at Mulago National Referral Hospital in Uganda. METHODS This was a prospective cohort study of 220 low birth weight neonates discharged from the Special Care Baby Unit at Mulago National Referral Hospital. These were followed up to 28 completed days of life, or death, whichever occurred first. Proportions were used to express mortality. To determine the predictors of mortality, Cox hazards regression was performed. RESULTS Of the 220 enrolled participants, 216 (98.1%) completed the follow-up. The mean gestational age of study participants was 34 ±3 weeks. The median weight at discharge was 1,650g (IQR: 1,315g -1,922g) and 46.1% were small for gestational age. During follow-up, 14/216 (6.5%) of neonates died. Mortality was highest (7/34, 20.6%) among neonates with discharge weights less than 1,200g. The causes of death included presumed neonatal sepsis (10/14, 71.4%), suspected aspiration pneumonia (2/14, 14.3%), and suspected cot death (2/14, 14.3%). The median time to death after discharge was 11 days (range 3-16 days). The predictors of mortality were a discharge weight of less than 1,200g (adj HR: 23.47, p <0.001), a 5-minute Apgar score of less than 7 (adj HR: 4.25, p = 0.016), and a diagnosis of neonatal sepsis during admission (adj HR: 7.93, p = 0.009). CONCLUSION Post-discharge mortality among low birth weight neonates at Mulago National Referral Hospital is high. A discharge weight of less than 1,200g may be considered unsafe among neonates. Caregiver education about neonatal danger signs, and measures to prevent sepsis, aspiration, and cot death should be emphasized before discharge and during follow-up visits.
Collapse
Affiliation(s)
- Ronald Nsubuga
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
- School Public Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Joseph Rujumba
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
- School Public Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Saleh Nyende
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Stevens Kisaka
- Department of Paediatrics and Child Health, Mulago National Referral Hospital, Kampala, Uganda
| | - Richard Idro
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
- School Public Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Jolly Nankunda
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
- School Public Health, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
22
|
Zhang SL, McGann CM, Duranova T, Strysko J, Steenhoff AP, Gezmu A, Nakstad B, Arscott-Mills T, Bayani O, Moorad B, Tlhako N, Richard-Greenblatt M, Planet PJ, Coffin SE, Silverman MA. Maternal and neonatal IgG against Klebsiella pneumoniae are associated with broad protection from neonatal sepsis: a case-control study of hospitalized neonates in Botswana. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.28.24308042. [PMID: 38854006 PMCID: PMC11160826 DOI: 10.1101/2024.05.28.24308042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Sepsis is the leading postnatal cause of neonatal mortality worldwide. Globally Klebsiella pneumoniae is the leading cause of sepsis in hospitalized neonates. This study reports development and evaluation of ELISA for anti-Klebsiella IgG using dried blood spot samples and evaluates the association of anti-Klebsiella IgG (anti-Kleb IgG) antibodies in maternal and neonatal samples and the risk of neonatal sepsis. Neonates and their mothers were enrolled at 0-96 hours of life in the neonatal unit of a tertiary referral hospital in Gaborone, Botswana and followed until death or discharge to assess for episodes of blood culture-confirmed neonatal sepsis. Neonates with sepsis had significantly lower levels of Kleb-IgG compared to neonates who did not develop sepsis (Mann-Whitney U, p=0.012). Similarly, samples from mothers of neonates who developed sepsis tended to have less Kleb-IgG compared to mothers of controls (p=0.06). The inverse correlation between Kleb-IgG levels and all-cause bacteremia suggests that maternal Kleb-IgG is broadly protective through cross-reactivity with common bacterial epitopes. These data support the continued use of immunoglobulin assays using DBS samples to explore the role of passive immunity on neonatal sepsis risk and reaffirm the critical need for research supporting the development of maternal vaccines for neonatal sepsis.
Collapse
Affiliation(s)
- Siqi Linsey Zhang
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Carolyn M McGann
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tereza Duranova
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan Strysko
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew P Steenhoff
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alemayehu Gezmu
- Faculties of Medicine & Health Sciences, Department of Paediatric & Adolescent Health, University of Botswana, Gaborone, Botswana
| | - Britt Nakstad
- Faculties of Medicine & Health Sciences, Department of Paediatric & Adolescent Health, University of Botswana, Gaborone, Botswana
| | - Tonya Arscott-Mills
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - One Bayani
- Faculties of Medicine & Health Sciences, Department of Paediatric & Adolescent Health, University of Botswana, Gaborone, Botswana
| | - Banno Moorad
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - Nametso Tlhako
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - Melissa Richard-Greenblatt
- Hospital for Sick Children, Toronto, Canada
- Department of Laboratory and Pathobiology, University of Toronto, Canada
| | - Paul J Planet
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan E Coffin
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A Silverman
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
23
|
Sabharwal V, Taglauer E, Demos R, Snyder-Cappione J, Shaik-Dasthagirisaheb YB, Parker-Kelleher S, Hunnewell J, Boateng J, Clarke K, Yuen R, Barnett ED, Wachman EM, Yarrington CD. Comparison of Anti-SARS-CoV-2-Specific Antibody Signatures in Maternal and Infant Blood after COVID-19 Infection versus COVID-19 Vaccination during Pregnancy. Am J Perinatol 2024; 41:e2970-e2977. [PMID: 37774748 DOI: 10.1055/a-2183-9109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
OBJECTIVE The Advisory Committee on Immunization Practices and The American College of Obstetricians and Gynecologists recommend coronavirus disease 2019 (COVID-19) vaccine for pregnant persons to prevent severe illness and death. The objective was to examine levels of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG, IgM, and IgA against spike protein receptor binding domain (RBD) and nucleocapsid protein (NCP) in maternal and infant/cord blood at delivery after COVID 19 vaccination compared with SARS-CoV-2 infection at in mother-infant dyads at specified time points. STUDY DESIGN Mothers with SARS-CoV-2 infection (n = 31) or COVID-19 vaccination (n = 25) during pregnancy were enrolled between July 2020 and November 2021. Samples were collected at delivery and IgG, IgM, and IgA to RBD of spike and NCPs compared in the infected and vaccinated groups. Timing of infection/vaccination prior to delivery and correlation with antibody levels was performed. RESULTS The majority of participants received vaccination within 90 days of delivery and over half received the Pfizer BioNTech vaccine. There were no significant correlations between antibody levels and timing of infection or vaccination. Infant IgG levels to the RBD domain of spike protein were higher in the vaccinated group (n = 25) as compared with the infants born to mothers with infection (n = 31). Vaccination against COVID-19 during pregnancy was associated with detectable maternal and infant anti-RBD IgG levels at delivery irrespective of the timing of vaccination. CONCLUSION Timing of vaccination had no correlation to the antibody levels suggesting that the timing of maternal vaccination in the cohort did not matter. There was no IgM detected in infants from vaccinated mothers. Infants from vaccinated mothers had robust IgG titers to RBD, which have a lasting protective effect in infants. KEY POINTS · COVID-19 vaccination during pregnancy had detectable antibody.. · No correlation between antibody levels and timing of vaccination.. · Infants from vaccinated mothers had robust IgG titers to RBD..
Collapse
Affiliation(s)
| | | | - Riley Demos
- Boston University School of Medicine, Boston, Massachusetts
| | | | | | | | | | - Jeffery Boateng
- Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | - Katherine Clarke
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | - Rachel Yuen
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | | | - Elisha M Wachman
- Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | | |
Collapse
|
24
|
Borghesi A. Life-threatening infections in human newborns: Reconciling age-specific vulnerability and interindividual variability. Cell Immunol 2024; 397-398:104807. [PMID: 38232634 DOI: 10.1016/j.cellimm.2024.104807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/19/2024]
Abstract
In humans, the interindividual variability of clinical outcome following exposure to a microorganism is immense, ranging from silent infection to life-threatening disease. Age-specific immune responses partially account for the high incidence of infection during the first 28 days of life and the related high mortality at population level. However, the occurrence of life-threatening disease in individual newborns remains unexplained. By contrast, inborn errors of immunity and their immune phenocopies are increasingly being discovered in children and adults with life-threatening viral, bacterial, mycobacterial and fungal infections. There is a need for convergence between the fields of neonatal immunology, with its in-depth population-wide characterization of newborn-specific immune responses, and clinical immunology, with its investigations of infections in patients at the cellular and molecular levels, to facilitate identification of the mechanisms of susceptibility to infection in individual newborns and the design of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Borghesi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia, EU, Italy; School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland.
| |
Collapse
|
25
|
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
|
26
|
Zhou YP, Li P, Zhang Y, Wang XZ, Yang B, Mei MJ, Chen S, Cheng H, Zhang W, Luo MH. A case of congenital human cytomegalovirus infection with placental and pulmonary calcification despite presence of intrauterine IgG. J Med Virol 2023; 95:e29327. [PMID: 38112155 DOI: 10.1002/jmv.29327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Affiliation(s)
- Yue-Peng Zhou
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Ping Li
- Xiangya Hospital, Central South University, Changsha, China
| | - Youming Zhang
- Xiangya Hospital, Central South University, Changsha, China
| | - Xian-Zhang Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bo Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Meng-Jie Mei
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shizhen Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Han Cheng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Weishe Zhang
- Xiangya Hospital, Central South University, Changsha, China
| | - Min-Hua Luo
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Burrack N, Adar A, Goldbart A, Weissmann S, Cohen B, Hazan I, Horev A, Golan-Tripto I. Monocyte and neutrophil to lymphocyte ratios in hospitalized children with RSV bronchiolitis. Pediatr Pulmonol 2023; 58:3530-3541. [PMID: 37728225 DOI: 10.1002/ppul.26687] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/14/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
OBJECTIVE Acute bronchiolitis is the most common cause of hospitalization in young children. Data on monocyte-to-lymphocyte-ratio (MLR) and neutrophil-to-lymphocyte-ratio (NLR) as biomarkers are limited. We aim to evaluate these ratios in children hospitalized with respiratory syncytial virus (RSV) bronchiolitis and their value as biomarkers for severe clinical outcomes. STUDY DESIGN A single-center retrospective cohort study of children aged <2 years hospitalized due to RSV bronchiolitis, between January 2018 and March 2022, with a complete blood count upon admission. We divided the cohort into quartiles based on MLR and NLR values. We examined associations between quartiles and four clinical severity outcomes. RESULTS A total of 2038 children (median age: 4.4 months, IQR: 1.9-9.8) were included in the study. The median MLR and NLR values for quartiles 1-4 were 0.14, 0.22, 0.30, 0.47, and 0.37, 0.70, 1.16, 2.29, respectively. Children with higher MLR had higher hospitalization rates to the pediatric intensive care unit (PICU) (Q1 2.4%, Q4 9.4%, p < .001), extended hospital stays (Q1 19.4%, Q4 32%, p < .001), and lower minimal oxygen saturation (Q1 90%, Q4 87%, p < .001). Cut-off values of 0.34 for MLR and 0.67 for NLR optimally identified PICU admissions. In a model accounting for age and sex, the fourth MLR quartile had an RR of 3.4 (95% CI: 1.76-7.22) and successfully predicted PICU admissions (area under the curve = 0.73; 95% CI: 0.681-0.789). CONCLUSIONS MLR and NLR are potential biomarkers for identifying children with RSV bronchiolitis at a higher risk for severe outcomes, specifically PICU admission.
Collapse
Affiliation(s)
- Nitzan Burrack
- Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Assaf Adar
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Pediatrics, Soroka University Medical Center, Beer-Sheva, Israel
| | - Aviv Goldbart
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Pediatrics, Soroka University Medical Center, Beer-Sheva, Israel
- Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Sarah Weissmann
- Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Bracha Cohen
- Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
| | - Itai Hazan
- Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Amir Horev
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Pediatric Dermatology Service, Soroka University Medical Center, Beer Sheva, Israel
| | - Inbal Golan-Tripto
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Pediatrics, Soroka University Medical Center, Beer-Sheva, Israel
- Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel
| |
Collapse
|
28
|
Willemsen JE, Borghans JA, Bont LJ, Drylewicz J. Maternal vaccination against RSV can substantially reduce childhood mortality in low-income and middle-income countries: A mathematical modeling study. Vaccine X 2023; 15:100379. [PMID: 37711264 PMCID: PMC10498305 DOI: 10.1016/j.jvacx.2023.100379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is a leading cause of childhood mortality in infants below 6 months of age. In low-income and middle-income countries (LMICs), the public health burden is substantial and resources are limited. It is critical to inform decision makers about effectiveness of new interventions. Methods We developed a mathematical model where individual RSV subtype A (RSV-A) and B (RSV-B) maternally derived neutralizing titers were predicted at time of birth after maternal vaccination with the RSV prefusion F protein-based vaccine. We estimated the subsequent duration of vaccine-induced immunity and compared this to the age at time of death distribution in the RSV GOLD Mortality Database to predict the potential impact of maternal vaccination on RSV-related childhood mortality. We used country-specific timing of antenatal care visits distributions and mortality estimates to make country-specific predictions for number of cases averted. Findings The model predicts that on average a neonate born at 40 weeks gestational age will be protected between 6 and 7 months from RSV-A and approximately 5 months from RSV-B related mortality. We estimated the potential impact of RSV-related mortality for in-hospital and out-of-hospital cases in LMICs and predicted that in 51 GAVI-eligible countries maternal vaccination could avert between 55% and 63% of the RSV-related in-hospital mortality cases below 6 months of age. Interpretation We show that maternal vaccination could substantially decrease RSV-A and RSV-B related in-hospital and out-of-hospital mortality in LMICs in the first 6 months of life.
Collapse
Affiliation(s)
- Joukje E. Willemsen
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Division of Infectious Diseases, Department of Pediatrics, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - José A.M. Borghans
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Louis J. Bont
- Division of Infectious Diseases, Department of Pediatrics, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Julia Drylewicz
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
29
|
Yasa B, Memur S, Ozturk DY, Bagci O, Uslu SI, Cetinkaya M. Neonatal Outcomes of Premature Infants Born to Women with the Novel Coronavirus (SARS-CoV-2) Infection: A Case Control Study. Am J Perinatol 2023; 40:1715-1724. [PMID: 34839474 DOI: 10.1055/s-0041-1740177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Novel coronavirus disease 2019 (COVID-19) is a disease associated with atypical pneumonia caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). The first cases of COVID-19 were reported in Wuhan at the end of 2019. Transmission usually occurs via infected droplets and close personal contact; the possibility of vertical transmission is still under debate. This retrospective study aimed to analyze clinical characteristics of premature infants born to mothers with symptomatic COVID-19 disease. STUDY DESIGN This case control study compared the clinical and laboratory data of 20 premature infants born to mothers infected with SARS-CoV-2 with sex and gestational age-matched historical controls. RESULTS The median gestational age and birth weight in both groups were similar. Respiratory distress developed in 11 (55.5%) infants in study group and 19 (47.5%) infants in control group. Mechanical ventilation and endotracheal surfactant administration rates were similar. Median duration of hospitalization was 8.5 (2-76) days in study group and 12 days in historical controls. Real-time reverse-transcription polymerase chain reaction tests (RT-PCR) of nasopharyngeal swab samples for SARS-CoV-2 were found to be negative twice, in the first 24 hours and later at 24 to 48 hours of life. No neutropenia or thrombocytopenia was detected in the study group. Patent ductus arteriosus, bronchopulmonary dysplasia, and necrotizing enterocolitis rates were similar between groups. No mortality was observed in both groups. CONCLUSION To the best of our knowledge, this is one of the few studies evaluating the clinical outcomes of premature infants born to SARS-CoV-2 infected mothers. There was no evidence of vertical transmission of SARS-CoV-2 from symptomatic SARS-CoV-2-infected women to the neonate in our cohort. The neonatal outcomes also seem to be favorable with no mortality in preterm infants. KEY POINTS · SARS-CoV-2 pandemic is a challenge for pregnant women.. · Neonatal outcomes of premature infants born to mothers infected with SARS-CoV-2 not well defined.. · SARS-CoV-2 infection seems to have no adverse effect on mortality and morbidity in premature infants..
Collapse
Affiliation(s)
- Beril Yasa
- Neonatology Department, Istanbul Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Seyma Memur
- Neonatology Department, Istanbul Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Dilek Y Ozturk
- Neonatology Department, Istanbul Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Onur Bagci
- Neonatology Department, Istanbul Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Sait I Uslu
- Neonatology Department, Istanbul Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Merih Cetinkaya
- Neonatology Department, Istanbul Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| |
Collapse
|
30
|
Bennike TB. Advances in proteomics: characterization of the innate immune system after birth and during inflammation. Front Immunol 2023; 14:1254948. [PMID: 37868984 PMCID: PMC10587584 DOI: 10.3389/fimmu.2023.1254948] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023] Open
Abstract
Proteomics is the characterization of the protein composition, the proteome, of a biological sample. It involves the large-scale identification and quantification of proteins, peptides, and post-translational modifications. This review focuses on recent developments in mass spectrometry-based proteomics and provides an overview of available methods for sample preparation to study the innate immune system. Recent advancements in the proteomics workflows, including sample preparation, have significantly improved the sensitivity and proteome coverage of biological samples including the technically difficult blood plasma. Proteomics is often applied in immunology and has been used to characterize the levels of innate immune system components after perturbations such as birth or during chronic inflammatory diseases like rheumatoid arthritis (RA) and inflammatory bowel disease (IBD). In cancers, the tumor microenvironment may generate chronic inflammation and release cytokines to the circulation. In these situations, the innate immune system undergoes profound and long-lasting changes, the large-scale characterization of which may increase our biological understanding and help identify components with translational potential for guiding diagnosis and treatment decisions. With the ongoing technical development, proteomics will likely continue to provide increasing insights into complex biological processes and their implications for health and disease. Integrating proteomics with other omics data and utilizing multi-omics approaches have been demonstrated to give additional valuable insights into biological systems.
Collapse
Affiliation(s)
- Tue Bjerg Bennike
- Medical Microbiology and Immunology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
31
|
Liu S, Wang P, Shi X, Weng T, Zhong J, Zhang X, Qu J, Chen L, Xu Q, Meng X, Xiong H, Wu D, Fang D, Peng B, Zhang D. Maternal antibody transfer rate of vaccination against SARS-CoV-2 before or during early pregnancy and its protective effectiveness on offspring. J Med Virol 2023; 95:e29125. [PMID: 37800607 DOI: 10.1002/jmv.29125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
This study focuses on maternal antibody transfer following vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) before or during early pregnancy and its potential protective effects on infants, providing scientific evidence for vaccination strategies. This prospective study tested the samples for SARS-CoV-2 IgG antibody titers and neutralizing capacity and tracked the infections after birth. Perform multivariate analysis of factors influencing antibody transfer rate, newborn antibody titers, and infant infection. Total 87.1% (122/140) women received coronavirus disease 2019 (COVID-19) vaccine before or during early pregnancy, and 28 of them had breakthrough infection. The maternal and neonatal IgG positive rates at delivery were 60.7% (85/140) and 60.8% (87/143), respectively. A positive correlation was found between neonatal and maternal IgG antibody titers. Compared with the median IgG antibody transfer rate of infected pregnant women, that of vaccinated but not infected pregnant women was higher (1.21 versus: 1.53 [two doses], 1.71 [three doses]). However, neonatal IgG antibodies were relatively low (174.91 versus: 0.99 [two doses], 8.18 [three doses]), and their neutralizing capacity was weak. The overall effectiveness of maternal vaccination in preventing infant infection was 27.0%, and three doses had higher effectiveness than two doses (64.3% vs. 19.6%). Multivariate analysises showed that in vaccination group women receiving three doses or in infection group women with longer interval between infection and delivery had a higher antibody transfer rate and neonatal IgG antibody titer. More than half of women vaccinated before or during early pregnancy can achieve effective antibody transfer to newborns. However, the neonatal IgG antibody titer is low and has a weak neutralizing capacity, providing limited protection to infants.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Ping Wang
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Xiaolu Shi
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Tingsong Weng
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Jiayi Zhong
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Xiaomin Zhang
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Jing Qu
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Long Chen
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Qing Xu
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xiang Meng
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Husheng Xiong
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Dawei Wu
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Dajun Fang
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Bo Peng
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Dingmei Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
32
|
Whitlock AE, Moskowitzova K, Labuz DF, Sewall N, Mullin K, Kycia I, Zurakowski D, Fauza DO. Fetal Secretory IgA Delivery via Transamniotic Fetal Immunotherapy (TRAFIT) in a Rodent Model. J Pediatr Surg 2023; 58:2050-2053. [PMID: 37029025 DOI: 10.1016/j.jpedsurg.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/16/2023] [Accepted: 03/10/2023] [Indexed: 04/09/2023]
Abstract
PURPOSE We sought to determine the feasibility and routing kinetics of transamniotic fetal delivery of secretory immunoglobulin-A (SIgA), in a rodent model. METHODS Fetuses (n = 94) from seven time-dated pregnant dams received intra-amniotic injections on gestational day 17 (E17, term = E21-22) of either saline (n = 15) or a solution of 1 mg/mL of ≥95% homogeneous human SIgA (n = 79). Animals were euthanized daily at E18-E21 for quantification of the IgA component by ELISA at gestational membranes, placenta, and select fetal anatomical sites against saline controls procured at term. Statistical analysis was by Mann-Whitney U-test. RESULTS None of the saline-injected animals had detectable human IgA. SIgA-injected fetuses showed human IgA in the stomach aspirate, intestinal wall, lungs, liver, and serum at all time points. IgA levels were significantly higher in the gastric aspirate and in the intestine than in all other sites (p < 0.001 for both), with intestinal levels remaining stable through E18-E21 (p = 0.09-0.62 pairwise). Serum and placental levels were consistently low throughout, reaching near zero levels by E21. CONCLUSIONS The chronology of exogenous secretory-IgA kinetics after intra-amniotic injection is suggestive of fetal uptake by ingestion, leading to consistent levels in the gastrointestinal tract. Transamniotic fetal immunotherapy (TRAFIT) with secretory-IgA may become a novel strategy for enhancing early mucosal immunity. LEVEL OF EVIDENCE N/A (animal and laboratory study). TYPE OF STUDY Animal and laboratory study.
Collapse
Affiliation(s)
- Ashlyn E Whitlock
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Kamila Moskowitzova
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Daniel F Labuz
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Nicole Sewall
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Katherine Mullin
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Ina Kycia
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Kayatani AKK, Bobbili N, Megnekou R, Matsunaga M, Leke RFG, Taylor DW. Are high avidity antibodies to Plasmodium falciparum antigens preferentially transferred across the placenta of premature and term babies? Placenta 2023; 140:11-19. [PMID: 37515848 PMCID: PMC10528449 DOI: 10.1016/j.placenta.2023.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 07/31/2023]
Abstract
INTRODUCTION Transplacental transport of maternal IgG via the neonatal Fc receptor (FcRn) provides babies with passive immunity. Several factors are reported to influence transport, including the avidity of antibodies (Abs) for their cognate antigens. Unfortunately, information on the role of antibody (Ab) avidity is limited. This study investigated if i) antibodies (Abs) with high avidity for 6 Plasmodium falciparum antigens and tetanus toxoid (TTx) were preferentially transferred to premature and term Cameroonian babies and ii) if Ab avidity was increased in babies whose mothers had placental malaria (PM), implicating the involvement of immune complexes. METHODS Total IgG (mg/ml) and Abs to malarial antigens (AMA1, EBA-175, MSP1-42, MSP2, MSP3, DBL5 of VAR2CSA) and TTx were measured in paired mother-cord samples obtained from premature and term deliveries in Cameroon. Half the women had PM at delivery. Avidity Indices (AIs) were determined by treating antigen-bound-Abs with different molar concentrations of NH4SCN and calculating 50% endpoints. RESULTS Total IgG and antigen-specific Abs increased in cord blood with gestational age; however, AIs did not. AIs in paired maternal-cord blood samples were strongly associated for all antigens (r = 0.77-0.96). However, no significant different in AIs was found between paired mother-cord blood samples for any of the antigens (p values > 0.05). Similarly, Ab avidity was not increased in cord blood of babies whose mothers had PM or hypergammaglobulinemia. DISCUSSION Overall, there was no evidence that higher avidity Abs to any of the malarial antigens or TTx were preferentially transferred to Cameroonian babies.
Collapse
Affiliation(s)
- Alexander K K Kayatani
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Bioscience Building, Room 320, Honolulu, HI, 96813, USA.
| | - Naveen Bobbili
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Bioscience Building, Room 320, Honolulu, HI, 96813, USA.
| | - Rosette Megnekou
- The Biotechnology Center, University of Yaoundé 1, BP 3851, Messa, Yaoundé, Cameroon.
| | - Masako Matsunaga
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Medical Education Building, Room 411, Honolulu, HI, 96813, USA.
| | - Rose F G Leke
- The Biotechnology Center, University of Yaoundé 1, BP 3851, Messa, Yaoundé, Cameroon.
| | - Diane Wallace Taylor
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Bioscience Building, Room 320, Honolulu, HI, 96813, USA.
| |
Collapse
|
34
|
Hernández-Caravaca I, Moros-Nicolás C, González-Brusi L, Romero de Ávila MJ, De Paco Matallana C, Pelegrín P, Castaño-Molina MÁ, Díaz-Meca L, Sánchez-Romero J, Martínez-Alarcón L, Avilés M, Izquierdo-Rico MJ. Colostrum Features of Active and Recovered COVID-19 Patients Revealed Using Next-Generation Proteomics Technique, SWATH-MS. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1423. [PMID: 37628421 PMCID: PMC10453012 DOI: 10.3390/children10081423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Colostrum performs nutritional, anti-inflammatory and anti-infective functions and promotes immune system formation and organ development. The new coronavirus, SARS-CoV-2, has generated concerns about viral transmission through human milk, with a lack of evidence about human milk's protective effects against the infection. This study aimed at analyzing presence of the virus and at identifying the protein expression profile of human colostrum in active and COVID-19-recovered patients. Colostrum samples were collected from women with COVID-19 (n = 3), women recently recovered from the infection (n = 4), and non-infected women (n = 5). The samples were analyzed by means of RT-qPCR to determine presence of the virus and using SWATH-MS for proteomic analysis. Proteomic results were then analyzed using bioinformatic methods. The viral tests were negative for SARS-CoV-2 in the colostrum from COVID-19 patients. The proteomic analysis identified 301 common proteins in all samples analyzed. Nineteen proteins were upregulated and 7 were downregulated in the COVID-19 group versus the control samples, whereas 18 were upregulated and 7 were downregulated when comparing the COVID-19 group to the recovered group. Eleven proteins were biomarkers of active COVID-19 infection. Ten were upregulated: ACTN1, CD36, FAM3B, GPRC5B, IGHA2, IGK, PLTP, RAC1, SDCBP and SERPINF1, and one was downregulated: PSAP. These proteins are mainly related to immunity, inflammatory response and protein transport. In conclusion, the results of this study suggest that colostrum is not a vehicle for mother-to-child SARS-CoV-2 transmission. Moreover, the colostrum's proteome of active and recuperated patients indicate that it could provide immune benefits to infants.
Collapse
Affiliation(s)
- Iván Hernández-Caravaca
- Department of Community Nursing, Preventive Medicine and Public Health and History of Science, Campus de Sant Vicent del Raspeig, University of Alicante, 03690 Alicante, Spain;
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
| | - Carla Moros-Nicolás
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Murcia, Campus Mare Nostrum (CMN), 30120 Murcia, Spain;
| | - Leopoldo González-Brusi
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Murcia, Campus Mare Nostrum (CMN), 30120 Murcia, Spain;
| | - Mª José Romero de Ávila
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Murcia, Campus Mare Nostrum (CMN), 30120 Murcia, Spain;
| | - Catalina De Paco Matallana
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Servicio de Obstetricia y Ginecología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain;
| | - Pablo Pelegrín
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Departamento de Bioquímica y Biología Molecular “B” e Inmunología, Facultad de Medicina, Universidad de Murcia, Campus Mare Nostrum (CMN), 30120 Murcia, Spain
| | - María Ángeles Castaño-Molina
- Servicio de Obstetricia y Ginecología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain;
- Departamento de Enfermería, Facultad de Enfermería, Universidad de Murcia, Campus Mare Nostrum (CMN), 30120 Murcia, Spain
| | - Lucía Díaz-Meca
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Servicio de Obstetricia y Ginecología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain;
| | - Javier Sánchez-Romero
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Servicio de Obstetricia y Ginecología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain;
| | - Laura Martínez-Alarcón
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Departamento de Enfermería, Facultad de Enfermería, Universidad de Murcia, Campus Mare Nostrum (CMN), 30120 Murcia, Spain
- Unit, Department of Surgery, Virgen de la Arrixaca University Hospital, 30120 Murcia, Spain
| | - Manuel Avilés
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Murcia, Campus Mare Nostrum (CMN), 30120 Murcia, Spain;
| | - Mª José Izquierdo-Rico
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120 Murcia, Spain; (C.M.-N.); (L.G.-B.); (C.D.P.M.); (P.P.); (L.D.-M.); (L.M.-A.); (M.A.)
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Murcia, Campus Mare Nostrum (CMN), 30120 Murcia, Spain;
| |
Collapse
|
35
|
Brebant D, Couffignal C, Manchon P, Duquesne S, Picone O, Vauloup-Fellous C. Transplacental transfer of anti-SARS-CoV-2 neutralizing antibodies in comparison to other pathogens total antibodies. J Clin Virol 2023; 165:105495. [PMID: 37295035 PMCID: PMC10212596 DOI: 10.1016/j.jcv.2023.105495] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/11/2023]
Abstract
BACKGROUNDS Due to immaturity of their immune system, passive maternal immunization is essential for newborns during their first months of life. Therefore, in the current context of intense circulation of SARS-CoV-2, identifying factors influencing the transfer ratio (TR) of neutralizing antibodies against SARS-CoV-2 (NAb) appears important. METHODS Our study nested in the COVIPREG cohort (NCT04355234), included mothers who had a SARS-CoV-2 PCR positive during their pregnancy and their newborns. Maternal and neonatal NAb levels were measured with the automated iFlash system. RESULTS For the 173 mother-infant pairs included in our study, the median gestational age (GA) at delivery was 39.4 weeks of gestation (WG), and 29.7 WG at maternal SARS-CoV-2 infection. Using a multivariate logistic model, having a NAb TR above 1 was positively associated with a longer delay from maternal positive SARS-CoV-2 PCR to delivery (aOR 1.09, 95% CI: 1.03 - 1.17) and with a later GA at delivery (aOR = 1.58, 95% CI: 1.09 - 2.52). It was negatively associated with being a male newborn (aOR 0.21, 95% CI: 0.07 - 0.59). In 3rd trimester SARS-CoV-2 infected mothers, NAb TR was inferior to VZV, toxoplasmosis, CMV, measle and rubella's TR. However, in 1st or 2nd trimester infected mothers, only measle TR was different from NAb TR. CONCLUSION Male newborn of mothers infected by SARS-CoV-2 during their pregnancy appear to have less protection against SARS-CoV-2 in their first months of life than female newborns. Measle TR was superior to NAb TR even in case of 1st or 2nd trimester maternal SARS-CoV-2 infection. Future studies are needed to investigate possible differences in transmission of NAb following infection vs vaccination and its impact on TR.
Collapse
MESH Headings
- SARS-CoV-2/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Maternal-Fetal Exchange/immunology
- Gestational Age
- Humans
- Male
- Female
- COVID-19/blood
- COVID-19/immunology
- COVID-19/prevention & control
- Delivery, Obstetric
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Pregnancy
- Infant, Newborn
- Sex Characteristics
- COVID-19 Vaccines
- Vaccination
- Pregnancy Complications/blood
- Pregnancy Complications/immunology
- Infant, Newborn, Diseases/immunology
- Infant, Newborn, Diseases/prevention & control
- Infectious Disease Transmission, Vertical/prevention & control
- Paris
- Adult
Collapse
Affiliation(s)
- Diane Brebant
- Université Paris-Saclay, Inserm U1193, AP-HP, Hôpital Paul Brousse, Virology department, Villejuif 94800, France; Groupe de Recherche sur les Infections pendant la Grossesse (GRIG), France.
| | - Camille Couffignal
- Université Paris Cité, INSERM CIC-EC 1425, AP-HP, Hôpital Bichat - Claude Bernard, Clinical Research Department, France
| | - Pauline Manchon
- Université Paris Cité, INSERM CIC-EC 1425, AP-HP, Hôpital Bichat - Claude Bernard, Clinical Research Department, France
| | - Sandra Duquesne
- Université Paris-Saclay, Inserm U1193, AP-HP, Hôpital Paul Brousse, Virology department, Villejuif 94800, France
| | - Olivier Picone
- Université Paris Cité, Hôpital louis Mourier, Obstetrical department, France; IAME U1137, Inserm, Université Paris Cité, France; Groupe de Recherche sur les Infections pendant la Grossesse (GRIG), France
| | - Christelle Vauloup-Fellous
- Université Paris-Saclay, Inserm U1193, AP-HP, Hôpital Paul Brousse, Virology department, Villejuif 94800, France; Groupe de Recherche sur les Infections pendant la Grossesse (GRIG), France.
| |
Collapse
|
36
|
Whitlock AE, Moskowitzova K, Kycia I, Zurakowski D, Fauza DO. Fetal Alloimmune Hemolytic Anemia (AHA) as a Potential Target for Transamniotic Fetal Immunotherapy (TRAFIT). J Pediatr Surg 2023; 58:1107-1110. [PMID: 36914465 DOI: 10.1016/j.jpedsurg.2023.02.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023]
Abstract
PURPOSE Fetal alloimmune hemolytic anemia (AHA) resulting from maternal antibodies against fetal erythrocytes may require fetal administration of immunoglobulin-G (IgG) via invasive methods. IgG can reach the fetal circulation after transamniotic fetal immunotherapy (TRAFIT). We sought to both develop a model of AHA and to test TRAFIT as a potential treatment. METHODS Sprague-Dawley fetuses (n = 113) received intra-amniotic injections on gestational-day 18 (E18, term = E21) of either saline (control; n = 40), anti-rat-erythrocyte antibodies (AHA; n = 37), or anti-rat-erythrocyte antibodies plus IgG (AHA + IgG; n = 36). At term, blood was procured for red blood count (RBC), hematocrit, or ELISA for inflammatory markers. RESULTS There was no difference in survival [95% (107/113)] across groups (p = 0.87). Both hematocrit and RBC were significantly lower in the AHA group than controls (p < 0.001). Although still significantly lower than controls (p < 0.001), both hematocrit and RBC significantly increased in AHA + IgG group compared to AHA alone (p < 0.001). Pro-inflammatory TNF-α and IL1-β were significantly elevated from controls in the AHA group, but not in AHA + IgG (p < 0.001-0.159). CONCLUSIONS Intra-amniotic injection of anti-rat-erythrocyte antibodies can reproduce manifestations of fetal AHA, constituting a practical model of this disease. Transamniotic fetal immunotherapy with IgG reduces anemia in this model and may emerge as a new minimally invasive means of treatment. TYPE OF STUDY Animal and laboratory study. LEVEL OF EVIDENCE N/A (animal and laboratory study).
Collapse
Affiliation(s)
- Ashlyn E Whitlock
- Departments of Surgery, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA
| | - Kamila Moskowitzova
- Departments of Surgery, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA
| | - Ina Kycia
- Departments of Surgery, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA
| | - David Zurakowski
- Departments of Surgery, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA
| | - Dario O Fauza
- Departments of Surgery, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Zelini P, d'Angelo P, Zavaglio F, Soleymaninejadian E, Mariani L, Perotti F, Dominoni M, Tonello S, Sainaghi P, Minisini R, Apostolo D, Lilleri D, Spinillo A, Baldanti F. Inflammatory and Immune Responses during SARS-CoV-2 Infection in Vaccinated and Non-Vaccinated Pregnant Women and Their Newborns. Pathogens 2023; 12:pathogens12050664. [PMID: 37242334 DOI: 10.3390/pathogens12050664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Pregnant women are more susceptible to severe disease associated with SARS-CoV-2 infection. We performed a prospective study to analyze the inflammatory and immune profile after SARS-CoV-2 infection occurring in vaccinated or non-vaccinated pregnant women and their newborns. METHODS Twenty-five pregnant women with SARS-CoV-2 infection were enrolled, and sixteen cord blood samples were obtained at delivery. RESULTS We observed that IL-1β, TNF-α, Eotaxin, MIB-1β, VEGF, IL-15, IL-2, IL-5, IL-9, IL-10 and IL-1ra levels were significantly higher in vaccinated than non-vaccinated mothers. Furthermore, the newborns of the vaccinated mothers produced higher levels of IL-7, IL-5 and IL-12 compared to the newborns of non-vaccinated mothers. Anti-Spike (S) IgG levels were significantly higher in all vaccinated mothers and their newborns compared to the non-vaccinated group. We found that 87.5% of vaccinated women and 66.6% of non-vaccinated women mounted an S-specific T-cell response quantified by ELISpot assay. Moreover, 75.0% of vaccinated mothers and 38.4% of non-vaccinated mothers showed S-specific CD4+ T-cell proliferative response. The T-helper subset response was restricted to CD4+ Th1 in both vaccinated and non-vaccinated women. CONCLUSION A higher level of cytokines, IgG antibodies and memory T cells was noted in the vaccinated women. Furthermore, the maternal IgG antibody trans-placental transfer occurred more frequently in vaccinated mothers and may protect the newborn.
Collapse
Affiliation(s)
- Paola Zelini
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Piera d'Angelo
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Federica Zavaglio
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Ehsan Soleymaninejadian
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Liliana Mariani
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Francesca Perotti
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Mattia Dominoni
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Stelvio Tonello
- Immunoreumatology Laboratory, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, 28100 Novara, Italy
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Pierpaolo Sainaghi
- Immunoreumatology Laboratory, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, 28100 Novara, Italy
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Rosalba Minisini
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daria Apostolo
- Immunoreumatology Laboratory, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, 28100 Novara, Italy
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniele Lilleri
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Arsenio Spinillo
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Fausto Baldanti
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
38
|
Rosenberg YJ, Ordonez T, Khanwalkar US, Barnette P, Pandey S, Backes IM, Otero CE, Goldberg BS, Crowley AR, Leib DA, Shapiro MB, Jiang X, Urban LA, Lees J, Hessell AJ, Permar S, Haigwood NL, Ackerman ME. Evidence for the Role of a Second Fc-Binding Receptor in Placental IgG Transfer in Nonhuman Primates. mBio 2023; 14:e0034123. [PMID: 36946726 PMCID: PMC10127586 DOI: 10.1128/mbio.00341-23] [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: 02/09/2023] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
Transplacental transfer of maternal antibodies provides the fetus and newborn with passive protection against infectious diseases. While the role of the highly conserved neonatal Fc receptor (FcRn) in transfer of IgG in mammals is undisputed, recent reports have suggested that a second receptor may contribute to transport in humans. We report poor transfer efficiency of plant-expressed recombinant HIV-specific antibodies, including engineered variants with high FcRn affinity, following subcutaneous infusion into rhesus macaques close to parturition. Unexpectedly, unlike those derived from mammalian tissue culture, plant-derived antibodies were essentially unable to cross macaque placentas. This defect was associated with poor Fcγ receptor binding and altered Fc glycans and was not recapitulated in mice. These results suggest that maternal-fetal transfer of IgG across the three-layer primate placenta may require a second receptor and suggest a means of providing maternal antibody treatments during pregnancy while avoiding fetal harm. IMPORTANCE This study compared the ability of several human HIV envelope-directed monoclonal antibodies produced in plants with the same antibodies produced in mammalian cells for their ability to cross monkey and mouse placentas. We found that the two types of antibodies have comparable transfer efficiencies in mice, but they are differentially transferred across macaque placentas, consistent with a two-receptor IgG transport model in primates. Importantly, plant-produced monoclonal antibodies have excellent binding characteristics for human FcRn receptors, permitting desirable pharmacokinetics in humans. The lack of efficient transfer across the primate placenta suggests that therapeutic plant-based antibody treatments against autoimmune diseases and cancer could be provided to the mother while avoiding transfer and preventing harm to the fetus.
Collapse
Affiliation(s)
| | - Tracy Ordonez
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Iara M. Backes
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Claire E. Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | | | - Andrew R. Crowley
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - David A. Leib
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mariya B. Shapiro
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | | | | | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
39
|
Lubrano C, Mancon A, Anelli GM, Gagliardi G, Corneo R, Bianchi M, Coco C, Dal Molin G, Vignali M, Schirripa I, Di Simone N, Pavone G, Pellegrino A, Gismondo MR, Savasi VM, Cetin I. Immune Response and Transplacental Antibody Transfer in Pregnant Women after COVID-19 Vaccination. J Pers Med 2023; 13:jpm13040689. [PMID: 37109075 PMCID: PMC10141882 DOI: 10.3390/jpm13040689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
COVID-19 infection is associated with increased risk of pregnancy complications, making vaccination during pregnancy critical for mother-neonate dyads. Few data, often with an unrepresentative sample size, are available on SARS-CoV-2 vaccine-induced humoral and cell-mediated response. Here, we evaluated anti-S antibody and interferon-gamma (IFN-γ) production elicited by SARS-CoV-2 immunization in maternal and neonatal plasma. Pregnant women (n = 230) were prospectively enrolled and classified as unvaccinated (n = 103) and vaccinated (n = 127); after serological screening for previous infections, assays were performed on 126 dyads, 15 mothers and 17 newborns. Positive anti-S antibodies were found in most of the vaccinated subjects, regardless of timespan between immunization and delivery (range: 7-391 days). A total of 89 of 92 vaccinated women showed a broad response to COVID-19 immunization and highly effective placental transfer, as attested by anti-S positive rates (maternal = 96.7%, cord = 96.6%). Most of our subjects had indeterminate results in an IGRA assay, preventing a conclusive evaluation of IFN-γ production. Indeed, pregnancy-related hormonal changes may influence T-cell response with an impact on IFN-γ production. Positive pregnancy and perinatal outcomes reinforce the evidence that the anti-SARS-CoV-2 immunization is effective and well-tolerated in pregnant women and also protective for the fetus/neonate, even though it was not possible to define the related IFN-γ production and role.
Collapse
Affiliation(s)
- Chiara Lubrano
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Alessandro Mancon
- Laboratory of Clinical Microbiology, Virology and Bioemergencies, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, 20157 Milan, Italy
| | - Gaia Maria Anelli
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Gloria Gagliardi
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Roberta Corneo
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Micol Bianchi
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Chiara Coco
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Giulia Dal Molin
- Department of Biomedical Science for Health, Macedonio Melloni Hospital-ASST Fatebenefratelli Sacco, University of Milan, 20133 Milan, Italy
| | - Michele Vignali
- Department of Biomedical Science for Health, Macedonio Melloni Hospital-ASST Fatebenefratelli Sacco, University of Milan, 20133 Milan, Italy
| | - Irene Schirripa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Giulia Pavone
- Department of Woman, Child and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy
- Unit of Obstetrics and Gynecology, Alessandro Manzoni Hospital, ASST Lecco, 23900 Lecco, Italy
| | - Antonio Pellegrino
- Unit of Obstetrics and Gynecology, Alessandro Manzoni Hospital, ASST Lecco, 23900 Lecco, Italy
| | - Maria Rita Gismondo
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
- Laboratory of Clinical Microbiology, Virology and Bioemergencies, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, 20157 Milan, Italy
| | - Valeria Maria Savasi
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
- Department of Woman, Child and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy
| | - Irene Cetin
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
- Department of Woman, Mother and Neonate, Buzzi Children's Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy
| |
Collapse
|
40
|
Rak K, Styczyńska M, Godyla-Jabłoński M, Bronkowska M. Some Immune Parameters of Term Newborns at Birth Are Associated with the Concentration of Iron, Copper and Magnesium in Maternal Serum. Nutrients 2023; 15:nu15081908. [PMID: 37111127 PMCID: PMC10141145 DOI: 10.3390/nu15081908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The nutritional status of pregnant women is considered to affect fetal development and the health condition of newborns, including their immune system. We investigated the relationship between the concentrations of magnesium (Mg), calcium (Ca), zinc (Zn), iron (Fe) and copper (Cu) in maternal serum (MS) and the concentrations of IgG antibodies and antineutrophil cytoplasmatic auto-antibodies against lactoferrin (Lf-ANCA) in umbilical cord serum (UCS). IgG was considered as a promoter of immunity, and Lf-ANCA as an inhibitor. The examined group consisted of 98 pregnant women and their healthy term newborn children. The concentrations of mineral elements were measured by FAAS/FAES, while the concentrations of antibodies were determined by ELISA. Excessive MS Fe and insufficient MS Cu were related to insufficient UCS IgG and excessive UCS Lf-ANCA. The correlation analysis showed confirming results. Adequate UCS IgG and Lf-ANCA were related to MS Mg at the strictly lower limit of the reference values. The results obtained seem to suggest that an excess of Fe and a deficiency of Cu in pregnancy may adversely affect some immune parameters of newborns. Reference values for MS Mg are likely to require reconsideration. It would be advisable to monitor the nutritional status of pregnant women with minerals in order to support the immune capacity of newborns.
Collapse
Affiliation(s)
- Karolina Rak
- Department of Human Nutrition, Faculty of Biotechnology and Food Science, Wroclaw University of Environmental and Life Sciences, 51-630 Wrocław, Poland
| | - Marzena Styczyńska
- Department of Human Nutrition, Faculty of Biotechnology and Food Science, Wroclaw University of Environmental and Life Sciences, 51-630 Wrocław, Poland
| | - Michaela Godyla-Jabłoński
- Department of Human Nutrition, Faculty of Biotechnology and Food Science, Wroclaw University of Environmental and Life Sciences, 51-630 Wrocław, Poland
| | - Monika Bronkowska
- Institute of Health Sciences, Collegium Salutis Humanae, University of Opole, 45-060 Opole, Poland
| |
Collapse
|
41
|
Transplacental transfer of SARS-CoV-2 antibodies: a cohort study. Eur J Clin Microbiol Infect Dis 2023; 42:277-285. [PMID: 36692603 PMCID: PMC9872727 DOI: 10.1007/s10096-023-04553-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023]
Abstract
The purpose of this study was to examine the transfer rate of SARS-CoV-2 IgG antibodies in pregnancy and newborns. Two Danish labor wards screened all women for SARS-CoV-2 by PCR upon arrival. Women (n = 99) with a SARS-CoV-2 PCR-positive nasopharyngeal (NP) swab or with a household member with a positive swab at labor or any time during pregnancy, or COVID-19 symptoms upon admission (November 2020 through August 2021), were included. Mother and infant were tested by NP swabs at delivery, and maternal and infant (umbilical cord) venous blood samples were collected. We obtained clinical information including previous PCR test results from the medical records. SARS-Cov-2 IgM and quantified IgG antibodies were measured by enzyme-linked immunosorbent assay and transfer ratios of IgG. We detected IgG antibodies in 73 women and 65 cord blood sera and found a strong correlation between SARS-CoV-2 IgG concentrations in maternal and umbilical cord sera (r = 0.9; p < 0.05). Transfer ratio was > 1.0 in 51 out of 73 (69%) infants and > 1.5 in 26 (35%). We found that transfer was proportional to time from a positive SARS-CoV-2 PCR NP swab to delivery (r = 0.5; p < 0.05). Transfer ratios of SARS-CoV-2 antibodies were associated with time from infection to delivery with transfer ratios of more than 1.0 in the majority of seropositive mother-infant dyads.
Collapse
|
42
|
Romero-Ibarguengoitia ME, Flores-Salazar ZL, Arroyo-García KD, Soto-Gámez R, Leal-Meléndez JA, René Garza-Herrera M, Bennett-Vidales G, Cabrera MH, González-Habib R, Jiménez LP, Garza-Bulnes R, Barco-Flores IA, Castillo-Figueroa LF, Garza-Silva A, Rivera-Cavazos A, Rivera-Salinas D, González-Cantú A, Sanz-Sánchez MÁ. Evaluation of Transplacental Antibody Transfer in Pregnant Women Immunized with Different SARS-CoV-2 Homologous or Heterologous Schemes. Vaccines (Basel) 2023; 11:vaccines11020415. [PMID: 36851294 PMCID: PMC9961240 DOI: 10.3390/vaccines11020415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
There is scarce information related to transplacental antibody transfer against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with different homologous and heterologous vaccination schemes. This study aimed to correlate the magnitude of transplacental transfer anti-SARS-CoV-2 antibodies in different homologous and heterologous schemes. An observational cross-sectional study was developed to identify pregnant women vaccinated against SARS-CoV-2. They were questioned about their immunization status; blood samples from the mother, umbilical cord during labor, and the newborn 72 h after birth were taken to measure anti-S1 and anti-S2 specific IgG antibodies for SARS-CoV-2. We recruited 104 women with a median age of 29 (SD 1.17). We found antibodies in all newborns with vaccinated mothers. Homologous BNT162b2 mRNA regimen had the highest mean (SD) antibody titers (AU/mL) in maternal (994.93 (3.08), p = 0.039), umbilical cord (1316.43 (2.79), p = 0.016), and newborn (1192.02 (3.55), p = 0.020) blood. The generalized linear model showed a positive effect over antibodies with at least one dose in maternal (β = -1.1, p = 0.002) and newborn (β= -0.717, p = 0.044) blood, and with two doses (β = -0.684, p = 0.026) in umbilical cord blood. In conclusion, antibodies were detected in all vaccinated women and their newborns. Transfer of antibodies was found from the first dose, and the levels increased with the number of vaccine doses. Vaccination should be encouraged in pregnant women with any available scheme.
Collapse
Affiliation(s)
- Maria Elena Romero-Ibarguengoitia
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Dirección de Enseñanza e Investigación en Salud, Hospital Christus Muguerza Sistema de Salud, Calle Miguel Hidalgo y Costilla 2525, Obispado, Monterrey 64000, Nuevo León, Mexico
- Correspondence: ; Tel.: +52-81-8865-5656
| | - Zulema Lourdes Flores-Salazar
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Departamento de Neonatología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | - Kimberly Dariela Arroyo-García
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Departamento de Ginecología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | - Rafael Soto-Gámez
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Departamento de Ginecología, Hospital Clínica Nova de Monterrey, Avenida del Bosque 139, Cuauhtémoc, Cuauhtémoc, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | | | | | - Gordon Bennett-Vidales
- Departamento de Neonatología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | - Mauricio Hurtado Cabrera
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Roberto González-Habib
- Departamento de Ginecología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
- Dirección de Enseñanza e Investigación en Salud, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | - Liliann Peña Jiménez
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Raúl Garza-Bulnes
- Departamento de Neonatología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
| | | | - Luis Fernando Castillo-Figueroa
- Departamento de Neonatología, Hospital Christus Muguerza Conchita, Calle 15 de Mayo 1822, María Luisa, Nuevo Obispado, Monterrey 64040, Nuevo León, Mexico
- Departamento de Neonatología, Hospital Clínica Nova de Monterrey, Avenida del Bosque 139, Cuauhtémoc, Cuauhtémoc, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Arnulfo Garza-Silva
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Andrea Rivera-Cavazos
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Diego Rivera-Salinas
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Arnulfo González-Cantú
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
- Especialidades Médicas, Escuela de Medicina, Universidad de Monterrey, Avenida Morones Prieto, 4500-Pte, Zona Valle Poniente, San Pedro Garza García 66238, Nuevo León, Mexico
- Dirección de Enseñanza e Investigación en Salud, Hospital Christus Muguerza Sistema de Salud, Calle Miguel Hidalgo y Costilla 2525, Obispado, Monterrey 64000, Nuevo León, Mexico
| | - Miguel Ángel Sanz-Sánchez
- Research Department, Hospital Clínica Nova de Monterrey, San Nicolás de los Garza 66450, Nuevo León, Mexico
| |
Collapse
|
43
|
Luo Y, Lv H, Zhao S, Sun Y, Liu C, Chen C, Liang W, Kwok KO, Teo QW, So RTY, Lin Y, Deng Y, Li B, Dai Z, Zhu J, Zhang D, Fernando J, Wu NC, Tun HM, Bruzzone R, Mok CKP, Mu X. Age-related seroprevalence trajectories of seasonal coronaviruses in children including neonates in Guangzhou, China. Int J Infect Dis 2023; 127:26-32. [PMID: 36481488 PMCID: PMC9721286 DOI: 10.1016/j.ijid.2022.11.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Four seasonal coronaviruses, including human coronavirus (HCoV)-229E and HCoV-OC43, HCoV-NL63, and HCoV-HKU1 cause approximately 15-30% of common colds in adults. However, the full landscape of the immune trajectory to these viruses that covers the whole childhood period is still not well understood. METHODS We evaluated the serological responses against the four seasonal coronaviruses in 1886 children aged under 18 years by using enzyme-linked immunosorbent assay. The optical density values against each HCoV were determined from each sample. Generalized additive models were constructed to determine the relationship between age and seroprevalence throughout the whole childhood period. The specific antibody levels against the four seasonal coronaviruses were also tested from the plasma samples of 485 pairs of postpartum women and their newborn babies. RESULTS The immunoglobulin (Ig) G levels of the four seasonal coronaviruses in the mother and the newborn babies were highly correlated (229E: r = 0.63; OC43: r = 0.65; NL63: r = 0.69; HKU1: r = 0.63). The seroprevalences in children showed a similar trajectory in that the levels of IgG in the neonates dropped significantly and reached the lowest level after the age of around 1 year (229E: 1.18 years; OC43: 0.97 years; NL63: 1.01 years; HKU1: 1.02 years) and then resurgence in the children who aged older than 1 year. Using the lowest level from the generalized additive models as our cutoff, the seroprevalences for HCoV-229E, HCoV-OC43, HCoV-NL63, and HCoV-HKU1 were 98.11%, 96.23%, 96.23% and 94.34% at the age of 16-18 years. CONCLUSION Mothers share HCoV-specific IgGs with their newborn babies and the level of maternal IgGs waned at around 1 year after birth. The resurgence of the HCoV-specific IgGs was found thereafter with the increase in age suggesting repeated infection occurred in children.
Collapse
Affiliation(s)
- Yasha Luo
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, China
| | - Huibin Lv
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Shilin Zhao
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Yuanxin Sun
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China,The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chengyi Liu
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, China
| | - Chunke Chen
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Weiwen Liang
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kin-on Kwok
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Wen Teo
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Ray TY So
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yihan Lin
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yuhong Deng
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, China
| | - Biyun Li
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, China
| | - Zixi Dai
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jie Zhu
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Dengwei Zhang
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Julia Fernando
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Nicholas C Wu
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, USA,Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, USA,Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, USA,Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Hein M. Tun
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China,The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Roberto Bruzzone
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chris KP Mok
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China,The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China,Corresponding authors:
| | - Xiaoping Mu
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, China,Corresponding authors:
| |
Collapse
|
44
|
Pereira RA, de Almeida VO, Vidori L, Colvero MO, Amantéa SL. Immunoglobulin G and subclasses placental transfer in fetuses and preterm newborns: a systematic review. J Perinatol 2023; 43:3-9. [PMID: 36209234 DOI: 10.1038/s41372-022-01528-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 02/07/2023]
Abstract
Immunoglobulin G (IgG) and subclasses are the only class of antibodies capable of crossing placenta and providing protection against neonatal infectious diseases, especially in premature infants. This systematic review aimed to review the literature concerning the transplacental transfer of IgG and its subclasses in fetuses and preterm newborns and compare the results with data from term neonates. Eleven studies were included in the final review. Most studies demonstrated a lower transplacental passage of IgG2 than other subclasses in term and preterm newborns and a more efficient passage of total IgG and IgG1 after 37 weeks of gestational age. These results elucidate the physiology of IgG subclass transfer during pregnancy and may explain one of the reasons preterm newborns are especially susceptible to specific pathogens, such as encapsulated bacteria.
Collapse
Affiliation(s)
| | | | - Lennon Vidori
- Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | | | | |
Collapse
|
45
|
Kono M, Iyo T, Murakami D, Sakatani H, Nanushaj D, Hotomi M. Maternal immunization with pneumococcal surface protein A provides the immune memories of offspring against pneumococcal infection. Front Cell Infect Microbiol 2023; 13:1059603. [PMID: 37033488 PMCID: PMC10076723 DOI: 10.3389/fcimb.2023.1059603] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Streptococcus pneumoniae (S. pneumoniae) is one of the most widespread pathogens in the world and one of the largest infectious causes of infant mortality. Although current vaccines have various benefits, antibiotic resistance and the inability to vaccinate infants less than one year old demands the development of new protective strategies. One strategy, 'maternal immunization', is to protect infants by passive immunity from an immunized mother, although its mechanism is still not fully understood. Materials and methods The current study aimed to acquire immunity against S. pneumoniae in infants by maternal immunization with pneumococcal common antigen, pneumococcal surface protein A (PspA). Four-week-old female mice were immunized with recombinant PspA intranasally twice a week for three weeks. Females were mated with age-matched males after immunization, and delivered offspring. Results The week-old offspring derived from and fostered by immunized mothers had more anti-PspA-specific antibody producing cells in the spleen than those derived from sham-immunized mothers. The offspring were raised up to four weeks old and were subcutaneously stimulated with recombinant PspA. The levels of anti-PspA IgG in sera after stimulation were significantly higher in the offspring derived from the immunized mothers and the induced specific antibody to PspA showed protective efficacy against systemic pneumococcal infection. Discussion Maternal immunization is suggested to be able to provide a sustained immune memory to offspring. The current study would be a milestone in the field of maternal immunization toward a universal pneumococcal vaccine.
Collapse
Affiliation(s)
- Masamitsu Kono
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Takuro Iyo
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Kinan Hospital, Tanabe, Japan
| | - Daichi Murakami
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Kinan Hospital, Tanabe, Japan
| | - Hideki Sakatani
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Denisa Nanushaj
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
- *Correspondence: Muneki Hotomi,
| |
Collapse
|
46
|
Creisher PS, Campbell AD, Perry JL, Roznik K, Burd I, Klein SL. Influenza subtype-specific maternal antibodies protect offspring against infection but inhibit vaccine-induced immunity and protection in mice. Vaccine 2022; 40:6818-6829. [PMID: 36253217 PMCID: PMC10024894 DOI: 10.1016/j.vaccine.2022.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/02/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Following influenza A virus (IAV) infection or vaccination during pregnancy, maternal antibodies are transferred to offspring in utero and during lactation. The age and sex of offspring may differentially impact the transfer and effects of maternal immunity on offspring. To evaluate the effects of maternal IAV infection on immunity in offspring, we intranasally inoculated pregnant mice with sublethal doses of mouse-adapted (ma) H1N1, maH3N2, or media (mock) at embryonic day 10. In offspring of IAV-infected dams, maternal subtype-specific antibodies peaked at postnatal day (PND) 23, remained detectable through PND 50, and were undetectable by PND 105 in both sexes. When offspring were challenged with homologous IAV at PND 23, both male and female offspring had greater clearance of pulmonary virus and less morbidity and mortality than offspring from mock-inoculated dams. Inactivated influenza vaccination (IIV) against homologous IAV at PND 23 caused lower vaccine-induced antibody responses and protection following live virus challenge in offspring from IAV than mock-infected dams, with this effect being more pronounced among female than male offspring. At PND 105, there was no impact of maternal infection status, but vaccination induced greater antibody responses and protection against challenge in female than male offspring of both IAV-infected and mock-inoculated dams. To determine if maternal antibody or infection interfered with vaccine-induced immunity and protection in early life, offspring were vaccinated and challenged against a heterosubtypic IAV (i.e., different IAV group than dam) at PND 23 or 105. Heterosubtypic IAV maternal immunity did not affect antibody responses after IIV or protection after live IAV challenge of vaccinated offspring at either age. Subtype-specific maternal IAV antibodies, therefore, provide protection independent of offspring sex but interfere with vaccine-induced immunity and protection in offspring with more pronounced effects among females than males.
Collapse
Affiliation(s)
- Patrick S Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ariana D Campbell
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jamie L Perry
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Katerina Roznik
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
47
|
Kayatani AKK, Leke RGF, Leke RIJ, Fogako J, Taylor DW. Transplacental transfer of total immunoglobulin G and antibodies to Plasmodium falciparum antigens between the 24th week of gestation and term. Sci Rep 2022; 12:18864. [PMID: 36344542 PMCID: PMC9640577 DOI: 10.1038/s41598-022-21908-8] [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: 08/05/2022] [Accepted: 10/05/2022] [Indexed: 11/09/2022] Open
Abstract
Full-term newborns have antibody (Ab) repertoires and levels similar to their mothers to help protect them from environmental pathogens. Unfortunately, preterm babies, especially those born < 34 weeks, have reduced levels of protective antibodies. In Africa, antibodies to Plasmodium falciparum are important in protection from malaria. This study investigated the transfer of total IgG and antibodies to 9 P. falciparum antigens and tetanus toxoid between 24 weeks and term. Paired maternal and cord samples from 166 preterm (24-37 weeks) and 154 term deliveries were used. Transfer efficiency was expressed as the ratio of Ab levels in cord to maternal plasma (CMR). At 24-25 weeks, CMR ranged from 0.31 to 0.94 for the different antigens; the rate of transfer was similar for all antigens between 24 and 40 weeks; resulting in median CMR of 0.49-0.95 at term. Babies of mothers with hypergammaglobulinemia and normal IgG levels had similar amounts of IgG, supporting data that saturation of the neonatal Fc-receptor occurs at ~ 16 mg IgG/ml. Thus, babies born prior to 34-35 weeks in Africa are likely to have reduced Ab levels to some, but not all antigens. Since IgG transfer is Fc-mediated, why differences exist in CMR among the antigens warrants further investigation.
Collapse
Affiliation(s)
- Alexander K. K. Kayatani
- grid.410445.00000 0001 2188 0957Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Bioscience Building, Honolulu, HI 96813 USA
| | - Rose G. F. Leke
- grid.412661.60000 0001 2173 8504Faculty of Medicine & Biomedical Sciences, The Biotechnology Center, University of Yaoundé 1, Messa, BP 3851, Yaoundé, Cameroon
| | - Robert I. J. Leke
- grid.412661.60000 0001 2173 8504Faculty of Medicine & Biomedical Sciences, The Biotechnology Center, University of Yaoundé 1, Messa, BP 3851, Yaoundé, Cameroon
| | - Josephine Fogako
- grid.412661.60000 0001 2173 8504Faculty of Medicine & Biomedical Sciences, The Biotechnology Center, University of Yaoundé 1, Messa, BP 3851, Yaoundé, Cameroon
| | - Diane Wallace Taylor
- grid.410445.00000 0001 2188 0957Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Bioscience Building, Honolulu, HI 96813 USA
| |
Collapse
|
48
|
Flannery DD, Gouma S, Dhudasia MB, Mukhopadhyay S, Pfeifer MR, Woodford EC, Briker SM, Triebwasser JE, Gerber JS, Morris JS, Weirick ME, McAllister CM, Hensley SE, Puopolo KM. Comparison of Maternal and Neonatal Antibody Levels After COVID-19 Vaccination vs SARS-CoV-2 Infection. JAMA Netw Open 2022; 5:e2240993. [PMID: 36350652 PMCID: PMC9647482 DOI: 10.1001/jamanetworkopen.2022.40993] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
Importance Pregnant persons are at an increased risk of severe COVID-19 from SARS-CoV-2 infection, and COVID-19 vaccination is currently recommended during pregnancy. Objective To ascertain the association of vaccine type, time from vaccination, gestational age at delivery, and pregnancy complications with placental transfer of antibodies to SARS-CoV-2. Design, Setting, and Participants This cohort study was conducted in Pennsylvania Hospital in Philadelphia, Pennsylvania, and included births at the study site between August 9, 2020, and April 25, 2021. Maternal and cord blood serum samples were available for antibody level measurements for maternal-neonatal dyads. Exposures SARS-CoV-2 infection vs COVID-19 vaccination. Main Outcomes and Measures IgG antibodies to the receptor-binding domain of the SARS-CoV-2 spike protein were measured by quantitative enzyme-linked immunosorbent assay. Antibody concentrations and transplacental transfer ratios were measured after SARS-CoV-2 infection or receipt of COVID-19 vaccines. Results A total of 585 maternal-newborn dyads (median [IQR] maternal age, 31 [26-35] years; median [IQR] gestational age, 39 [38-40] weeks) with maternal IgG antibodies to SARS-CoV-2 detected at the time of delivery were included. IgG was detected in cord blood from 557 of 585 newborns (95.2%). Among 169 vaccinated persons without SARS-CoV-2 infection, the interval from first dose of vaccine to delivery ranged from 12 to 122 days. The geometric mean IgG level among 169 vaccine recipients was significantly higher than that measured in 408 persons after infection (33.88 [95% CI, 27.64-41.53] arbitrary U/mL vs 2.80 [95% CI, 2.50-3.13] arbitrary U/mL). Geometric mean IgG levels were higher after vaccination with the mRNA-1273 (Moderna) vaccine compared with the BNT162b2 (Pfizer/BioNTech) vaccine (53.74 [95% CI, 40.49-71.33] arbitrary U/mL vs 25.45 [95% CI, 19.17-33.79] arbitrary U/mL; P < .001). Placental transfer ratios were lower after vaccination compared with after infection (0.80 [95% CI, 0.68-0.93] vs 1.06 [95% CI, 0.98-1.14]; P < .001) but were similar between the mRNA vaccines (mRNA-1273: 0.70 [95% CI, 0.55-0.90]; BNT162b2: 0.85 [95% CI, 0.69-1.06]; P = .25). Time from infection or vaccination to delivery was associated with transfer ratio in models that included gestational age at delivery and maternal hypertensive disorders, diabetes, and obesity. Placental antibody transfer was detectable as early as 26 weeks' gestation. Transfer ratio that was higher than 1.0 was present for 48 of 51 (94.1%) births at 36 weeks' gestation or later by 8 weeks after vaccination. Conclusions and Relevance This study found that maternal and cord blood IgG antibody levels were higher after COVID-19 vaccination compared with after SARS-CoV-2 infection, with slightly lower placental transfer ratios after vaccination than after infection. The findings suggest that time from infection or vaccination to delivery was the most important factor in transfer efficiency.
Collapse
Affiliation(s)
- Dustin D. Flannery
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Miren B. Dhudasia
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Madeline R. Pfeifer
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Emily C. Woodford
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sara M. Briker
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | | | - Jeffrey S. Gerber
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Jeffrey S. Morris
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Madison E. Weirick
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | | | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Karen M. Puopolo
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
49
|
Dowling DJ, Barman S, Smith AJ, Borriello F, Chaney D, Brightman SE, Melhem G, Brook B, Menon M, Soni D, Schüller S, Siram K, Nanishi E, Bazin HG, Burkhart DJ, Levy O, Evans JT. Development of a TLR7/8 agonist adjuvant formulation to overcome early life hyporesponsiveness to DTaP vaccination. Sci Rep 2022; 12:16860. [PMID: 36258023 PMCID: PMC9579132 DOI: 10.1038/s41598-022-20346-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Infection is the most common cause of mortality early in life, yet the broad potential of immunization is not fully realized in this vulnerable population. Most vaccines are administered during infancy and childhood, but in some cases the full benefit of vaccination is not realized in-part. New adjuvants are cardinal to further optimize current immunization approaches for early life. However, only a few classes of adjuvants are presently incorporated in vaccines approved for human use. Recent advances in the discovery and delivery of Toll-like receptor (TLR) agonist adjuvants have provided a new toolbox for vaccinologists. Prominent among these candidate adjuvants are synthetic small molecule TLR7/8 agonists. The development of an effective infant Bordetella pertussis vaccine is urgently required because of the resurgence of pertussis in many countries, contemporaneous to the switch from whole cell to acellular vaccines. In this context, TLR7/8 adjuvant based vaccine formulation strategies may be a promising tool to enhance and accelerate early life immunity by acellular B. pertussis vaccines. In the present study, we optimized (a) the formulation delivery system, (b) structure, and (c) immunologic activity of novel small molecule imidazoquinoline TLR7/8 adjuvants towards human infant leukocytes, including dendritic cells. Upon immunization of neonatal mice, this TLR7/8 adjuvant overcame neonatal hyporesponsiveness to acellular pertussis vaccination by driving a T helper (Th)1/Th17 biased T cell- and IgG2c-skewed humoral response to a licensed acellular vaccine (DTaP). This potent immunization strategy may represent a new paradigm for effective immunization against pertussis and other pathogens in early life.
Collapse
Affiliation(s)
- David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Soumik Barman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Alyson J Smith
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59802, USA
- Seagen, Bothell, WA, USA
| | - Francesco Borriello
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, 80131, Italy
- WAO Center of Excellence, Naples, 80131, Italy
- Generate Biomedicines, Cambridge, MA, USA
| | - Danielle Chaney
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Spencer E Brightman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
| | - Gandolina Melhem
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
| | - Byron Brook
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Manisha Menon
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
| | - Dheeraj Soni
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Simone Schüller
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Karthik Siram
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Hélène G Bazin
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59802, USA
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - David J Burkhart
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59802, USA
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT & Harvard, Cambridge, MA, USA.
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59802, USA.
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA.
| |
Collapse
|
50
|
Kochanowicz JF, Nowicka A, Al-Saad SR, Karbowski LM, Gadzinowski J, Szpecht D. Catheter-related bloodstream infections in infants hospitalized in neonatal intensive care units: a single center study. Sci Rep 2022; 12:13679. [PMID: 35953522 PMCID: PMC9372030 DOI: 10.1038/s41598-022-17820-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 08/01/2022] [Indexed: 11/09/2022] Open
Abstract
Sepsis in neonates carries a high morbidity and mortality rate and is among the most feared complications in the neonatal intensive care unit (NICU). Catheter-related bloodstream infections (CRBSI) are a common etiology of late-onset sepsis. The aim of this study was to compare risk factors and characteristics between patients according to the type of catheter that was utilized and according to birth weight classification. The study included 51 newborns with confirmed CRBSI, which were hospitalized in our level 3 NICU between January 2017 and December 2018. The study population was stratified according to the type of venous catheter utilized (peripherally inserted central catheter, central venous catheter (CVC), and peripheral venous catheter). Infants with low birth weight and those who required prolonged parenteral nutrition were most likely to develop CRBSI in our study group. The type of venous catheter was not associated with blood culture results. Also, infants with a birth weight of < 1500 g and > 1500 g did not differ in sepsis etiology. Further research is required to assess venous catheters relative risk of causing sepsis and if the outcome can be traced back specifically to catheter type or patient characteristics.
Collapse
Affiliation(s)
| | - Agnieszka Nowicka
- Chair and Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | - Janusz Gadzinowski
- Chair and Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Dawid Szpecht
- Chair and Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|