1
|
Singh VR, O'Donnell LA. Age-Stratified Treg Responses During Viral Infections of the Central Nervous System: A Literature Review. J Med Virol 2025; 97:e70315. [PMID: 40178106 PMCID: PMC11967158 DOI: 10.1002/jmv.70315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/24/2024] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Regulatory T cells (Tregs) play a vital role in limiting inflammation and resolving the immune response after a viral infection. Within the central nervous system (CNS), Tregs are especially important for the protection of neurons, which have limited regenerative capacity, and the preservation of myelin sheaths, which support neuronal function and survival. Nevertheless, viral infections of the CNS often result in enduring neurological dysfunction, especially in more vulnerable age groups such as newborns and the elderly. Although it is appreciated that Treg activity changes with age, it is unclear how these age-dependent changes impact viral CNS infections. In this review, we explore Treg development over the life of the host and discuss evidence for age-dependent Treg responses to peripheral viral infections. We also discuss the CNS-specific roles of Tregs, where both immunomodulatory and neuroprotective functions can contribute to preservation of brain cells. Finally, we examine the current evidence for Treg activity in neurotropic infections in the context of age, and highlight gaps in our understanding of Treg function in younger and older hosts. Overall, a better understanding of age-dependent Treg activity in the CNS may reveal opportunities for therapeutic interventions tailored to the most vulnerable ages.
Collapse
Affiliation(s)
- Vivek R. Singh
- School of Pharmacy and the Graduate School of Pharmaceutical SciencesDuquesne UniversityPittsburghPennsylvaniaUSA
| | - Lauren A. O'Donnell
- School of Pharmacy and the Graduate School of Pharmaceutical SciencesDuquesne UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
2
|
Tang MH, Ligthart I, Varga S, Lebeer S, van Overveld FJ, Rijkers GT. Mutual Interactions Between Microbiota and the Human Immune System During the First 1000 Days of Life. BIOLOGY 2025; 14:299. [PMID: 40136555 PMCID: PMC11940030 DOI: 10.3390/biology14030299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/25/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
The development of the human immune system starts during the fetal period in a largely, but probably not completely, sterile environment. During and after birth, the immune system is exposed to an increasingly complex microbiota. The first microbiota encountered during passage through the birth canal colonize the infant gut and induce the tolerance of the immune system. Transplacentally derived maternal IgG as well as IgA from breast milk protect the infant from infections during the first 100 days, during which the immune system further develops and immunological memory is formed. The Weaning and introduction of solid food expose the immune system to novel (food) antigens and allow for other microbiota to colonize. The cells and molecules involved in the mutual and intricate interactions between microbiota and the developing immune system are now beginning to be recognized. These include bacterial components such as polysaccharide A from Bacteroides fragilis, as well as bacterial metabolites such as the short-chain fatty acid butyrate, indole-3-aldehyde, and indole-3-propionic acid. All these, and probably more, bacterial metabolites have specific immunoregulatory functions which shape the development of the human immune system during the first 1000 days of life.
Collapse
Affiliation(s)
- Muy Heang Tang
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Ishbel Ligthart
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Samuel Varga
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Sarah Lebeer
- Lab of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerpen, Belgium;
| | - Frans J. van Overveld
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Ger T. Rijkers
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| |
Collapse
|
3
|
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
|
4
|
Paul A, Bui T, Muelbert M, Toldi G. T lymphocytes in human milk: Their role in immune system maturation through maternal microchimerism. J Reprod Immunol 2025; 168:104451. [PMID: 39951899 DOI: 10.1016/j.jri.2025.104451] [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: 10/22/2024] [Revised: 01/08/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Breastfeeding promotes the trafficking of immune cells and soluble factors from the mother to the neonate during lactation, resulting in maternal microchimerism. Human milk is abundant in T lymphocytes, but little is known about their priming and actions in neonatal mucosal tissues and their role in conferring immune tolerance in early life. This review summarises recent findings on the characteristics of human milk T cells compared to their counterparts in maternal and neonatal blood. We discuss how bioactive components of human milk, such as cytokines, hormones, and miRNA, may modulate the immune suppressive function of this cell subset. We shed light on the presence and possible functions of regulatory T cells (Tregs) in the breastfeeding triad of mother, human milk, and neonate, and how this subset of T lymphocytes may contribute to the prevention of immune pathologies, such as allergies and autoimmune diseases, later in life through human milk-induced maternal microchimerism in the newborn.
Collapse
Affiliation(s)
- Ayamita Paul
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Tram Bui
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Mariana Muelbert
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Gergely Toldi
- Liggins Institute, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
5
|
Tisseyre M, Collier M, Beeker N, Kaguelidou F, Treluyer JM, Chouchana L. Prenatal Exposure to Proton Pump Inhibitors and Risk of Serious Infections in Offspring During the First Year of Life: A Nationwide Cohort Study. Drug Saf 2025; 48:265-277. [PMID: 39630354 DOI: 10.1007/s40264-024-01496-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 02/16/2025]
Abstract
INTRODUCTION AND OBJECTIVE Proton pump inhibitor (PPI) use in children increases the risk of infections, prompting inquiry into the impact of prenatal PPIs exposure on serious infections in offspring. As a research gap in this area exists, this study aimed to address it by assessing the association between prenatal PPIs exposure and serious infections in infants during their first year of life. METHODS Using the French health insurance data warehouse (SNDS) (2013-2018), we conducted a retrospective cohort study on singleton, full-term liveborn non-immunocompromised infants, stratified by PPI use during the first three months of life (early-life use). Proton pump inhibitor dispensing in ambulatory care settings during pregnancy defined exposure. Outcomes concerned any serious infections in offspring aged between 3 and 12 months. Adjusted odds ratios (aORs) were estimated using logistic regression with multivariable models to control for potential confounders. RESULTS Of the 2,485,545 infants included, 497,060 (23.3%) were prenatally exposed to PPIs and 97,767 (4.6%) had PPI use during the first three months of life. Prenatal PPI exposure was associated with serious infections in offspring (aOR, 1.09 [95% CI, 1.07-1.10]) in infants without early-life PPIs use. No association was found for infants with early-life PPI use (aOR, 1.05 [95% CI, 1.00-1.11]). Gastrointestinal infections were the sole site with persistent significance. CONCLUSION Prenatal PPI exposure is common and is not associated with a major risk of serious infections in infants during their first year. However, even after adjusting for several confounding factors, a weak association remains, especially in infants without early-life PPI use. While offering reassurance, adherence to clinical guidelines is still crucial.
Collapse
Affiliation(s)
- Mylène Tisseyre
- Centre Régional de Pharmacovigilance, Service de Pharmacologie périnatale, pédiatrique et adulte, Hopital Cochin, Assistance Publique-Hopitaux de Paris (AP-HP), 27, rue du Faubourg Saint Jacques, 75014, Paris, France.
- UMR7323 « Pharmacologie et évaluation des thérapeutiques chez l'enfant et la femme enceinte », INSERM, Université Paris Cité, Paris, France.
| | - Mathis Collier
- UMR7323 « Pharmacologie et évaluation des thérapeutiques chez l'enfant et la femme enceinte », INSERM, Université Paris Cité, Paris, France
- Unité de Recherche clinique, Hopital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Nathanaël Beeker
- UMR7323 « Pharmacologie et évaluation des thérapeutiques chez l'enfant et la femme enceinte », INSERM, Université Paris Cité, Paris, France
- Unité de Recherche clinique, Hopital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Florentia Kaguelidou
- UMR7323 « Pharmacologie et évaluation des thérapeutiques chez l'enfant et la femme enceinte », INSERM, Université Paris Cité, Paris, France
- Centre d'Investigations Cliniques, INSERM CIC1426, Hôpital Robert Debré, APHP. Nord, Paris, France
| | - Jean-Marc Treluyer
- Centre Régional de Pharmacovigilance, Service de Pharmacologie périnatale, pédiatrique et adulte, Hopital Cochin, Assistance Publique-Hopitaux de Paris (AP-HP), 27, rue du Faubourg Saint Jacques, 75014, Paris, France
- UMR7323 « Pharmacologie et évaluation des thérapeutiques chez l'enfant et la femme enceinte », INSERM, Université Paris Cité, Paris, France
- Unité de Recherche clinique, Hopital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Laurent Chouchana
- Centre Régional de Pharmacovigilance, Service de Pharmacologie périnatale, pédiatrique et adulte, Hopital Cochin, Assistance Publique-Hopitaux de Paris (AP-HP), 27, rue du Faubourg Saint Jacques, 75014, Paris, France
- UMR7323 « Pharmacologie et évaluation des thérapeutiques chez l'enfant et la femme enceinte », INSERM, Université Paris Cité, Paris, France
| |
Collapse
|
6
|
Faas MM, Smink AM. Shaping immunity: the influence of the maternal gut bacteria on fetal immune development. Semin Immunopathol 2025; 47:13. [PMID: 39891756 PMCID: PMC11787218 DOI: 10.1007/s00281-025-01039-8] [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: 06/06/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025]
Abstract
The development of the fetal immune response is a highly complex process. In the present review, we describe the development of the fetal immune response and the role of the maternal gut bacteria in this process. In contrast to the previous belief that the fetal immune response is inert, it is now thought that the fetal immune response is uniquely tolerant to maternal and allo-antigens, but able to respond to infectious agents, such as bacteria. This is accomplished by the development of T cells toward regulatory T cells rather than toward effector T cells, but also by the presence of functional innate immune cells, such as monocytes and NK cells. Moreover, in fetuses there is different programming of CD8 + T cells and memory T cells toward innate immune cells rather than to adaptive immune cells. The maternal gut bacteria are important in shaping the fetal immune response by producing bacterial products and metabolites that pass the placenta into the fetus and influence development of the fetal immune response. Insight into how and when these products affect the fetal immune response may open new treatment options with pre- or probiotics to affect the maternal gut bacteria and therewith the fetal immune response.
Collapse
Affiliation(s)
- Marijke M Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen and University of Groningen, Hanzeplein 1, Groningen, 9713 GZ, The Netherlands.
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen and University of Groningen, Hanzeplein 1, Groningen, 9713 GZ, The Netherlands
| |
Collapse
|
7
|
Gomez-Salinero JM, Redmond D, Rafii S. Microenvironmental determinants of endothelial cell heterogeneity. Nat Rev Mol Cell Biol 2025:10.1038/s41580-024-00825-w. [PMID: 39875728 DOI: 10.1038/s41580-024-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
During development, endothelial cells (ECs) undergo an extraordinary specialization by which generic capillary microcirculatory networks spanning from arteries to veins transform into patterned organotypic zonated blood vessels. These capillary ECs become specialized to support the cellular and metabolic demands of each specific organ, including supplying tissue-specific angiocrine factors that orchestrate organ development, maintenance of organ-specific functions and regeneration of injured adult organs. Here, we illustrate the mechanisms by which microenvironmental signals emanating from non-vascular niche cells induce generic ECs to acquire specific inter-organ and intra-organ functional attributes. We describe how perivascular, parenchymal and immune cells dictate vascular heterogeneity and capillary zonation, and how this system is maintained through tissue-specific signalling activated by vasculogenic and angiogenic factors and deposition of matrix components. We also discuss how perturbation of organotypic vascular niche cues lead to erasure of EC signatures, contributing to the pathogenesis of disease processes. We also describe approaches that use reconstitution of tissue-specific signatures of ECs to promote regeneration of damaged organs.
Collapse
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Du Q, Liu X, Zhang R, Hu G, Liu Q, Wang R, Ma W, Hu Y, Fan Z, Li J. Placental and Fetal Microbiota in Rhesus Macaque: A Case Study Using Metagenomic Sequencing. Am J Primatol 2025; 87:e23718. [PMID: 39716039 DOI: 10.1002/ajp.23718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/04/2024] [Accepted: 11/30/2024] [Indexed: 12/25/2024]
Abstract
Recent evidence challenging the notion of a sterile intrauterine environment has sparked research into the origins and effects of fetal microbiota on immunity development during gestation. Rhesus macaques (RMs) serve as valuable nonhuman primate models due to their similarities to humans in development, placental structure, and immune response. In this study, metagenomic analysis was applied to the placenta, umbilical cord, spleen, gastrointestinal tissues of an unborn RM fetus, and the maternal intestine, revealing the diversity and functionality of microbes in these tissues. Additionally, gut metagenomic data of adult Rhesus macaques from our previous study, along with data from a human fetus obtained from public databases, were included for comparison. We observed substantial microbial sharing between the mother and fetus, with the microbial composition of the placenta and umbilical cord more closely resembling that of the fetal organs than the maternal intestine. Notably, compared with other adult RMs, there was a clear convergence between maternal and fetal microbiota, alongside distinct differences between the microbiota of adults and the fetus, which underscores the unique microbial profiles in fetal environments. Furthermore, the fetal microbiota displayed a less developed carbohydrate metabolism capacity than adult RMs. It also shared antibiotic resistance genes with both maternal and adult RM microbiomes, indicating potential vertical transmission. Comparative analysis of the metagenomes between the RM fetus and a human fetus revealed significant differences in microbial composition and genes, yet also showed similarities in certain abundant microbiota. Collectively, our results contribute to a more comprehensive understanding of the intrauterine microbial environment in macaques.
Collapse
Affiliation(s)
- Qiao Du
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Xu Liu
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Rusong Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Gang Hu
- SCU-SGHB Joint Laboratory on Nonhuman Primates Research, Sichuan Green-house Biotech Co. Ltd., Meishan, China
| | - Qinghua Liu
- SCU-SGHB Joint Laboratory on Nonhuman Primates Research, Sichuan Green-house Biotech Co. Ltd., Meishan, China
| | - Rui Wang
- SCU-SGHB Joint Laboratory on Nonhuman Primates Research, Sichuan Green-house Biotech Co. Ltd., Meishan, China
| | - Wen Ma
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Ying Hu
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Zhenxin Fan
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Jing Li
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Ma J, Wu H, Ma Z, Wu Z. Bacterial and host factors involved in zoonotic Streptococcal meningitis. Microbes Infect 2025; 27:105335. [PMID: 38582147 DOI: 10.1016/j.micinf.2024.105335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
Zoonotic streptococci cause several invasive diseases with high mortality rates, especially meningitis. Numerous studies elucidated the meningitis pathogenesis of zoonotic streptococci, some specific to certain bacterial species. In contrast, others are shared among different bacterial species, involving colonization and invasion of mucosal barriers, survival in the bloodstream, breaching the blood-brain and/or blood-cerebrospinal fluid barrier to access the central nervous system, and triggering inflammation of the meninges. This review focuses on the recent advancements in comprehending the molecular and cellular events of five major zoonotic streptococci responsible for causing meningitis in humans or animals, including Streptococcus agalactiae, Streptococcus equi subspecies zooepidemicus, Streptococcus suis, Streptococcus dysgalactiae, and Streptococcus iniae. The underlying mechanism was summarized into four themes, including 1) bacterial survival in blood, 2) brain microvascular endothelial cell adhesion and invasion, 3) penetration of the blood-brain barrier, and 4) activation of the immune system and inflammatory reaction within the brain. This review may contribute to developing therapeutics to prevent or mitigate injury of streptococcal meningitis and improve risk stratification.
Collapse
Affiliation(s)
- Jiale Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China; WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Huizhen Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China; WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Zhe Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China; WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China; Guangdong Provincial Key Laboratory of Research on the Technology of Pig-breeding and Pig-disease Prevention, Guangzhou 511400, China.
| |
Collapse
|
10
|
Rowell J, Lau CI, Ross S, Yanez DC, Peña OA, Chain B, Crompton T. Distinct T-cell receptor (TCR) gene segment usage and MHC-restriction between foetal and adult thymus. eLife 2024; 13:RP93493. [PMID: 39636212 PMCID: PMC11620746 DOI: 10.7554/elife.93493] [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: 12/07/2024] Open
Abstract
Here, we sequenced rearranged TCRβ and TCRα chain sequences in CD4+CD8+ double positive (DP), CD4+CD8- single positive (SP4) and CD4-CD8+ (SP8) thymocyte populations from the foetus and young adult mouse. We found that life-stage had a greater impact on TCRβ and TCRα gene segment usage than cell-type. Foetal repertoires showed bias towards 3'TRAV and 5'TRAJ rearrangements in all populations, whereas adult repertoires used more 5'TRAV gene segments, suggesting that progressive TCRα rearrangements occur less frequently in foetal DP cells. When we synchronised young adult DP thymocyte differentiation by hydrocortisone treatment the new recovering DP thymocyte population showed more foetal-like 3'TRAV and 5'TRAJ gene segment usage. In foetus we identified less influence of MHC-restriction on α-chain and β-chain combinatorial VxJ usage and CDR1xCDR2 (V region) usage in SP compared to adult, indicating weaker impact of MHC-restriction on the foetal TCR repertoire. The foetal TCRβ repertoire was less diverse, less evenly distributed, with fewer non-template insertions, and all foetal populations contained more clonotypic expansions than adult. The differences between the foetal and adult thymus TCR repertoires are consistent with the foetal thymus producing αβT-cells with properties and functions that are distinct from adult T-cells: their repertoire is less governed by MHC-restriction, with preference for particular gene segment usage, less diverse with more clonotypic expansions, and more closely encoded by genomic sequence.
Collapse
Affiliation(s)
- Jasmine Rowell
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Ching-In Lau
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Susan Ross
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Diana C Yanez
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Oscar A Peña
- School of Biochemistry, University of BristolBristolUnited Kingdom
| | - Benny Chain
- Division of Infection and Immunity, University College LondonLondonUnited Kingdom
| | - Tessa Crompton
- UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| |
Collapse
|
11
|
Olasunkanmi OI, Aremu J, Wong ML, Licinio J, Zheng P. Maternal gut-microbiota impacts the influence of intrauterine environmental stressors on the modulation of human cognitive development and behavior. J Psychiatr Res 2024; 180:307-326. [PMID: 39488009 DOI: 10.1016/j.jpsychires.2024.10.028] [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: 07/18/2023] [Revised: 11/01/2023] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
This review examines the longstanding debate of nature and intrauterine environmental challenges that shapes human development and behavior, with a special focus on the influence of maternal prenatal gut microbes. Recent research has revealed the critical role of the gut microbiome in human neurodevelopment, and evidence suggest that maternal microbiota can impact fetal gene and microenvironment composition, as well as immunophysiology and neurochemical responses. Furthermore, intrauterine neuroepigenetic regulation may be influenced by maternal microbiota, capable of having long-lasting effects on offspring behavior and cognition. By examining the complex relationship between maternal prenatal gut microbes and human development, this review highlights the importance of early-life environmental factors in shaping neurodevelopment and cognition.
Collapse
Affiliation(s)
- Oluwatayo Israel Olasunkanmi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| | - John Aremu
- Department of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Ma-Li Wong
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA
| | - Julio Licinio
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA.
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Migliorini A, Ge S, Atkins MH, Oakie A, Sambathkumar R, Kent G, Huang H, Sing A, Chua C, Gehring AJ, Keller GM, Notta F, Nostro MC. Embryonic macrophages support endocrine commitment during human pancreatic differentiation. Cell Stem Cell 2024; 31:1591-1611.e8. [PMID: 39406230 DOI: 10.1016/j.stem.2024.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/02/2024] [Accepted: 09/12/2024] [Indexed: 11/10/2024]
Abstract
Organogenesis is a complex process that relies on a dynamic interplay between extrinsic factors originating from the microenvironment and tissue-specific intrinsic factors. For pancreatic endocrine cells, the local niche consists of acinar and ductal cells as well as neuronal, immune, endothelial, and stromal cells. Hematopoietic cells have been detected in human pancreas as early as 6 post-conception weeks, but whether they play a role during human endocrinogenesis remains unknown. To investigate this, we performed single-nucleus RNA sequencing (snRNA-seq) of the second-trimester human pancreas and identified a wide range of hematopoietic cells, including two distinct subsets of tissue-resident macrophages. Leveraging this discovery, we developed a co-culture system of human embryonic stem cell-derived endocrine-macrophage organoids to model their interaction in vitro. Here, we show that macrophages support the differentiation and viability of endocrine cells in vitro and enhance tissue engraftment, highlighting their potential role in tissue engineering strategies for diabetes.
Collapse
Affiliation(s)
- Adriana Migliorini
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada.
| | - Sabrina Ge
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Michael H Atkins
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | | | - Gregory Kent
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Haiyang Huang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Angel Sing
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Conan Chua
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Adam J Gehring
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Faiyaz Notta
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
13
|
Giannoni E, Sanchez Sanchez G, Verdebout I, Papadopoulou M, Rezwani M, Ahmed R, Ladell K, Miners KL, McLaren JE, Fraser DJ, Price DA, Eberl M, Agyeman PKA, Schlapbach LJ, Vermijlen D. Sepsis shapes the human γδ TCR repertoire in an age- and pathogen-dependent manner. Eur J Immunol 2024; 54:e2451190. [PMID: 39072722 DOI: 10.1002/eji.202451190] [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: 04/15/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
Sepsis affects 25 million children per year globally, leading to 2.9 million deaths and substantial disability in survivors. Extensive characterization of interactions between the host and bacteria in children is required to design novel preventive and therapeutic strategies tailored to this age group. Vγ9Vδ2 T cells are the first T cells generated in humans. These cells are defined by the expression of Vγ9Vδ2 T-cell receptors (TCRs, using the TRGV9 and TRDV2 gene segments), which react strongly against the prototypical bacterial phosphoantigen HMBPP. We investigated this reactivity by analyzing the TCR δ (TRD) repertoire in the blood of 76 children (0-16 years) with blood culture-proven bacterial sepsis caused by HMBPP-positive Escherichia coli or by HMBPP-negative Staphylococcus aureus or by HMBPP-negative Streptococcus pneumoniae. Strikingly, we found that S. aureus, and to a lesser extent E. coli but not S. pneumoniae, shaped the TRDV2 repertoire in young children (<2 years) but not in older children or adults. This dichotomy was due to the selective expansion of a fetal TRDV2 repertoire. Thus, young children possess fetal-derived Vγ9Vδ2 T cells that are highly responsive toward specific bacterial pathogens.
Collapse
Affiliation(s)
- Eric Giannoni
- Clinic of Neonatology, Department Mother-Woman-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Isoline Verdebout
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Maria Papadopoulou
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Moosa Rezwani
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Raya Ahmed
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Kristin Ladell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Kelly L Miners
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - James E McLaren
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Donald J Fraser
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
- Wales Kidney Research Unit, Heath Park Campus, Cardiff, UK
- Directorate of Nephrology and Transplantation, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - David A Price
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Philipp K A Agyeman
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Luregn J Schlapbach
- Department of Intensive Care and Neonatology, and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
14
|
Guo A, Ludvigsson J, Lerchova T, Imberg H, Størdal K, Mårild K. Association Between Maternal Infections in Pregnancy and the Risk of Inflammatory Bowel Disease in the Offspring: Findings From Two Scandinavian Birth Cohorts. Inflamm Bowel Dis 2024:izae209. [PMID: 39248793 DOI: 10.1093/ibd/izae209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND The association of infections and antibiotic use in pregnancy and the risk of inflammatory bowel disease (IBD) development in the offspring have been scarcely investigated. We examined infection and antibiotic use in pregnancy and the risk of IBD in offspring. METHODS We followed participants from the All Babies in Southeast Sweden (ABIS) and the Norwegian mother father and child cohort (MoBa) from birth (1997-2009) until 2020-2021. IBD diagnosis was classified as ≥2 records in national registers. Information on infections (any, gastrointestinal, and respiratory), their timing (early or late in pregnancy), and antibiotic use in pregnancy were collected from questionnaires. Cox proportional-hazard regression and meta-analytic methods were used to estimate pooled adjusted hazard ratios (aHRs) for IBD and its subtypes, adjusted for parental IBD, maternal smoking, and education. Sensitivity analyses accounted for exposure to antibiotics and infections 0-12 months of age. RESULTS We followed 117 493 children for 2 024 299 person-years (follow-up 22.3 years in ABIS and 16.4 years in MoBa), including 451 IBD cases. The aHRs for any infection and respiratory infections in pregnancy and offspring IBD were close to one (aHR = 0.99 [95% CI = 0.73-1.33] and aHR = 1.00 [95% CI = 0.81-1.23], respectively). However, any versus no infection in early pregnancy was associated with IBD development (aHR = 1.26 [95% CI = 1.02-1.55]), particularly Crohn's disease (CD; aHR = 1.40 [95% CI = 1.01-1.93]). Any versus no gastrointestinal infection in late pregnancy was associated with offspring CD (aHR = 1.95 [95% CI = 1.34-2.84]). Antibiotic use in pregnancy was not associated with IBD in the child (aHR = 1.15 [95% CI = 0.93-1.44]). CONCLUSIONS In this binational birth cohort study, the risk of offspring IBD varied by infection type and timing but not with maternal antibiotic use in pregnancy.
Collapse
Affiliation(s)
- Annie Guo
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital, Region Östergötland, Linköping, Sweden
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Tereza Lerchova
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Imberg
- Statistiska Konsultgruppen, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ketil Størdal
- Children's Center, Oslo University Hospital, Oslo, Norway
- Department of Pediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karl Mårild
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Queen Silvia Children's Hospital, Gothenburg, Sweden
| |
Collapse
|
15
|
Reyes JGA, Ni D, Santner-Nanan B, Pinget GV, Kraftova L, Ashhurst TM, Marsh-Wakefield F, Wishart CL, Tan J, Hsu P, King NJC, Macia L, Nanan R. A unique human cord blood CD8 +CD45RA +CD27 +CD161 + T-cell subset identified by flow cytometric data analysis using Seurat. Immunology 2024; 173:106-124. [PMID: 38798051 DOI: 10.1111/imm.13803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
Advances in single-cell level analytical techniques, especially cytometric approaches, have led to profound innovation in biomedical research, particularly in the field of clinical immunology. This has resulted in an expansion of high-dimensional data, posing great challenges for comprehensive and unbiased analysis. Conventional manual analysis is thus becoming untenable to handle these challenges. Furthermore, most newly developed computational methods lack flexibility and interoperability, hampering their accessibility and usability. Here, we adapted Seurat, an R package originally developed for single-cell RNA sequencing (scRNA-seq) analysis, for high-dimensional flow cytometric data analysis. Based on a 20-marker antibody panel and analyses of T-cell profiles in both adult blood and cord blood (CB), we showcased the robust capacity of Seurat in flow cytometric data analysis, which was further validated by Spectre, another high-dimensional cytometric data analysis package, and conventional manual analysis. Importantly, we identified a unique CD8+ T-cell population defined as CD8+CD45RA+CD27+CD161+ T cell that was predominantly present in CB. We characterised its IFN-γ-producing and potential cytotoxic properties using flow cytometry experiments and scRNA-seq analysis from a published dataset. Collectively, we identified a unique human CB CD8+CD45RA+CD27+CD161+ T-cell subset and demonstrated that Seurat, a widely used package for scRNA-seq analysis, possesses great potential to be repurposed for cytometric data analysis. This facilitates an unbiased and thorough interpretation of complicated high-dimensional data using a single analytical pipeline and opens a novel avenue for data-driven investigation in clinical immunology.
Collapse
Affiliation(s)
- Julen Gabirel Araneta Reyes
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Nepean Hospital, Nepean Blue Mountains Local Health District, Penrith, New South Wales, Australia
- Nepean Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Nepean Hospital, Nepean Blue Mountains Local Health District, Penrith, New South Wales, Australia
- Nepean Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Brigitte Santner-Nanan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Nepean Hospital, Nepean Blue Mountains Local Health District, Penrith, New South Wales, Australia
- Nepean Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Gabriela Veronica Pinget
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Nepean Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Lucie Kraftova
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Nepean Clinical School, The University of Sydney, Sydney, New South Wales, Australia
- Department of Microbiology, Faculty of Medicine, University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
- Biomedical Center, Faculty of Medicine, Charles University, Pilsen, Czech Republic
| | - Thomas Myles Ashhurst
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, The University of Sydney and Centenary Institute, Sydney, New South Wales, Australia
| | - Felix Marsh-Wakefield
- Liver Injury and Cancer Program, Centenary Institute, Sydney, New South Wales, Australia
- Human Cancer and Viral Immunology Laboratory, The University of Sydney, Sydney, New South Wales, Australia
| | - Claire Leana Wishart
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Viral immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Ramaciotti Facility for Human System Biology, The University of Sydney and Centenary Institute, Sydney, New South Wales, Australia
| | - Jian Tan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Peter Hsu
- Kids Research, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Nicholas Jonathan Cole King
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, The University of Sydney and Centenary Institute, Sydney, New South Wales, Australia
- Viral immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Ramaciotti Facility for Human System Biology, The University of Sydney and Centenary Institute, Sydney, New South Wales, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Nano, The University of Sydney, Sydney, New South Wales, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, The University of Sydney and Centenary Institute, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ralph Nanan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Nepean Hospital, Nepean Blue Mountains Local Health District, Penrith, New South Wales, Australia
- Nepean Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
16
|
Moreno MDL, González-Rovira M, Martínez-Pancorbo C, Martín-Cameán M, Nájar-Moyano AM, Romero M, de la Hoz E, López-Beltrán C, Mellado E, Bartha JL, Brodin P, Rodríguez-Herrera A, Sainz-Bueno JA, Sousa C. Foetal gluten immunogenic peptides during pregnancy: a new determinant on the coeliac exposome. BMC Med 2024; 22:295. [PMID: 39020299 PMCID: PMC11256569 DOI: 10.1186/s12916-024-03495-9] [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: 03/03/2024] [Accepted: 06/20/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND The increasing incidence of coeliac disease is leading to a growing interest in active search for associated factors, even the intrauterine and early life. The exposome approach to disease encompasses a life course perspective from conception onwards has recently been highlighted. Knowledge of early exposure to gluten immunogenic peptides (GIP) in utero could challenge the chronology of early prenatal tolerance or inflammation, rather than after the infant's solid diet after birth. METHODS We developed an accurate and specific immunoassay to detect GIP in amniotic fluid (AF) and studied their accumulates, excretion dynamics and foetal exposure resulting from AF swallowing. One hundred twenty-five pregnant women with different gluten diets and gestational ages were recruited. RESULTS GIP were detectable in AF from at least the 16th gestational week in gluten-consuming women. Although no significant differences in GIP levels were observed during gestation, amniotic GIP late pregnancy was not altered by maternal fasting, suggesting closed-loop entailing foetal swallowing of GIP-containing AF and subsequent excretion via the foetal kidneys. CONCLUSIONS The study shows evidence, for the first time, of the foetal exposure to gluten immunogenic peptides and establishes a positive correlation with maternal gluten intake. The results obtained point to a novel physiological concept as they describe a plausible closed-loop circuit entailing foetal swallowing of GIP contained in AF and its subsequent excretion through the foetal kidneys. The study adds important new information to understanding the coeliac exposome.
Collapse
Affiliation(s)
- María de Lourdes Moreno
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - María González-Rovira
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | | | - María Martín-Cameán
- Obstetric and Gynecology Department, High Risk Pregnancy Unit, Autoimmune Diseases and Pregnancy Clinic, University Hospital La Paz, Madrid, Spain
| | - Ana María Nájar-Moyano
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Mercedes Romero
- Obstetric and Gynecology Department, Sagrado Corazón Hospital, Seville, Spain
| | - Esther de la Hoz
- Obstetric and Gynecology Department, Sagrado Corazón Hospital, Seville, Spain
| | | | - Encarnación Mellado
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - José Luis Bartha
- Obstetric and Gynecology Department, High Risk Pregnancy Unit, Autoimmune Diseases and Pregnancy Clinic, University Hospital La Paz, Madrid, Spain
| | - Petter Brodin
- Department of Women's and Children's Health, Karolinska Institutet, Uppsala, Sweden
| | - Alfonso Rodríguez-Herrera
- St. Luke's General Hospital, UCD School of Medicine, University College Dublin, Kilkenny &, Kilkenny, Ireland
| | - José Antonio Sainz-Bueno
- Department of Obstetrics and Gynecology, Valme University Hospital, Seville, Spain
- Department of Obstetrics and Gynecology, University of Seville, Seville, Spain
| | - Carolina Sousa
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| |
Collapse
|
17
|
Pirker AL, Vogl T. Development of systemic and mucosal immune responses against gut microbiota in early life and implications for the onset of allergies. FRONTIERS IN ALLERGY 2024; 5:1439303. [PMID: 39086886 PMCID: PMC11288972 DOI: 10.3389/falgy.2024.1439303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024] Open
Abstract
The early microbial colonization of human mucosal surfaces is essential for the development of the host immune system. Already during pregnancy, the unborn child is prepared for the postnatal influx of commensals and pathogens via maternal antibodies, and after birth this protection is continued with antibodies in breast milk. During this critical window of time, which extends from pregnancy to the first year of life, each encounter with a microorganism can influence children's immune response and can have a lifelong impact on their life. For example, there are numerous links between the development of allergies and an altered gut microbiome. However, the exact mechanisms behind microbial influences, also extending to how viruses influence host-microbe interactions, are incompletely understood. In this review, we address the impact of infants' first microbial encounters, how the immune system develops to interact with gut microbiota, and summarize how an altered immune response could be implied in allergies.
Collapse
Affiliation(s)
| | - Thomas Vogl
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Larbi A. From Genesis to Old Age: Exploring the Immune System One Cell at a Time with Flow Cytometry. Biomedicines 2024; 12:1469. [PMID: 39062042 PMCID: PMC11275137 DOI: 10.3390/biomedicines12071469] [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: 05/12/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The immune system is a highly complex and tightly regulated system that plays a crucial role in protecting the body against external threats, such as pathogens, and internal abnormalities, like cancer cells. It undergoes development during fetal stages and continuously learns from each encounter with pathogens, allowing it to develop immunological memory and provide a wide range of immune protection. Over time, after numerous encounters and years of functioning, the immune system can begin to show signs of erosion, which is commonly named immunosenescence. In this review, we aim to explore how the immune system responds to initial encounters with antigens and how it handles persistent stimulations throughout a person's lifetime. Our understanding of the immune system has greatly benefited from advanced technologies like flow cytometry. In this context, we will discuss the valuable contribution of flow cytometry in enhancing our knowledge of the immune system behavior in aging, with a specific focus on T-cells. Moreover, we will expand our discussion to the flow cytometry-based assessment of extracellular vesicles, a recently discovered communication channel in biology, and their implications for immune system functioning.
Collapse
Affiliation(s)
- Anis Larbi
- Medical and Scientific Affairs, Beckman Coulter Life Sciences, 22 Avenue des Nations, 93420 Villepinte, France;
- Department of Medicine, Division of Geriatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| |
Collapse
|
19
|
Clements RL, Kennedy EA, Song D, Campbell A, An HH, Amses KR, Miller-Ensminger T, Addison MM, Eisenlohr LC, Chou ST, Jurado KA. Human erythroid progenitors express antigen presentation machinery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601047. [PMID: 39005276 PMCID: PMC11244935 DOI: 10.1101/2024.06.27.601047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Early-life immune exposures can profoundly impact lifelong health. However, functional mechanisms underlying fetal immune development remain incomplete. Erythrocytes are not typically considered active immune mediators, primarily because erythroid precursors discard their organelles as they mature, thus losing the ability to alter gene expression in response to stimuli. Erythroid progenitors and precursors circulate in human fetuses and neonates. Although there is limited evidence that erythroid precursors are immunomodulatory, our understanding of the underlying mechanisms remains inadequate. To define the immunobiological role of fetal and perinatal erythroid progenitors and precursors, we analyzed single cell RNA-sequencing data and found that transcriptomics support erythroid progenitors as putative immune mediators. Unexpectedly, we discovered that human erythroid progenitors constitutively express Major Histocompatibility Complex (MHC) class II antigen processing and presentation machinery, which are hallmarks of specialized antigen presenting immune cells. Furthermore, we demonstrate that erythroid progenitors internalize and cleave foreign proteins into peptide antigens. Unlike conventional antigen presenting cells, erythroid progenitors express atypical costimulatory molecules and immunoregulatory cytokines that direct the development of regulatory T cells, which are critical for establishing maternal-fetal tolerance. Expression of MHC II in definitive erythroid progenitors begins during the second trimester, coinciding with the appearance of mature T cells in the fetus, and is absent in primitive progenitors. Lastly, we demonstrate physical and molecular interaction potential of erythroid progenitors and T cells in the fetal liver. Our findings shed light on a unique orchestrator of fetal immunity and provide insight into the mechanisms by which erythroid cells contribute to host defense.
Collapse
|
20
|
Nagafuji M, Fujiyama S, Ishii R, Shime M, Kitatsu T, Hoshino Y, Kanai Y, Arai J, Miyazono Y, Takada H. Effect of maturation at birth on the clinical features of neonatal cow's milk protein allergy: A retrospective study. J Pediatr Gastroenterol Nutr 2024; 79:48-53. [PMID: 38356291 DOI: 10.1002/jpn3.12157] [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: 07/25/2023] [Revised: 10/30/2023] [Accepted: 12/23/2023] [Indexed: 02/16/2024]
Abstract
Neonatal immune regulation transitions from fetal immunity and varies with maturation status, but its role in neonatal cow's milk protein allergy (CMPA) remains unknown. We studied the association between maturation status at birth and neonatal CMPA. Clinical and laboratory data of neonates presenting with CMPA symptoms were retrospectively collected from two tertiary hospitals. Patients were assessed according to gestational age at birth: preterm, late-preterm, and full-term. Fifty-five infants (26 females, 14 preterm, 15 late-preterm, and 26 full-term) were included; 44 were negative for milk-specific immunoglobulin E. Neonatal CMPA was common during moderately premature periods. Preterm infants exhibited longer latency from initial CM exposure to disease onset, lower incidence of bloody stool, and absence of elevated monocyte counts. However, immunoreactivity to CM antigens was retained in all infants. Neonatal CMPA features varied with infant maturation status at birth. Our results improve the understanding of intestinal immunity development, fetal/neonatal immune regulation, and CMPA pathogenesis.
Collapse
Affiliation(s)
- Motomichi Nagafuji
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| | - Satoshi Fujiyama
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| | - Ryota Ishii
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Michiru Shime
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| | - Toshinori Kitatsu
- Department of Pediatric Allergy, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| | - Yusuke Hoshino
- Department of Neonatology, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| | - Yu Kanai
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
- Department of Child Health, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Junichi Arai
- Department of Neonatology, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| | - Yayoi Miyazono
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
- Department of Child Health, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hidetoshi Takada
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
- Department of Child Health, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
21
|
Amissah HA, Combs SE, Shevtsov M. Tumor Dormancy and Reactivation: The Role of Heat Shock Proteins. Cells 2024; 13:1087. [PMID: 38994941 PMCID: PMC11240553 DOI: 10.3390/cells13131087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Tumors are a heterogeneous group of cell masses originating in various organs or tissues. The cellular composition of the tumor cell mass interacts in an intricate manner, influenced by humoral, genetic, molecular, and tumor microenvironment cues that dictate tumor growth or suppression. As a result, tumors undergo a period of a dormant state before their clinically discernible stage, which surpasses the clinical dormancy threshold. Moreover, as a genetically imprinted strategy, early-seeder cells, a distinct population of tumor cells, break off to dock nearby or extravasate into blood vessels to secondary tissues, where they form disseminated solitary dormant tumor cells with reversible capacity. Among the various mechanisms underlying the dormant tumor mass and dormant tumor cell formation, heat shock proteins (HSPs) might play one of the most important roles in how the dormancy program plays out. It is known that numerous aberrant cellular processes, such as malignant transformation, cancer cell stemness, tumor invasion, metastasis, angiogenesis, and signaling pathway maintenance, are influenced by the HSPs. An accumulating body of knowledge suggests that HSPs may be involved in the angiogenic switch, immune editing, and extracellular matrix (ECM) remodeling cascades, crucial genetically imprinted strategies important to the tumor dormancy initiation and dormancy maintenance program. In this review, we highlight the biological events that orchestrate the dormancy state and the body of work that has been conducted on the dynamics of HSPs in a tumor mass, as well as tumor cell dormancy and reactivation. Additionally, we propose a conceptual framework that could possibly underlie dormant tumor reactivation in metastatic relapse.
Collapse
Affiliation(s)
- Haneef Ahmed Amissah
- Institute of Life Sciences and Biomedicine, Department of Medical Biology and Medical Biology, FEFU Campus, Far Eastern Federal University, 690922 Vladivostok, Russia;
- Diagnostics Laboratory Department, Trauma and Specialist Hospital, CE-122-2486, Central Region, Winneba P.O. Box 326, Ghana
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
| | - Maxim Shevtsov
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| |
Collapse
|
22
|
Felício R, Franco A, Corrêa-Moreira D, Martinez de Carvalho F, Guimarães A, Vargas FR. Humoral immune transcriptome signature in myelomeningocele patients. J Reprod Immunol 2024; 163:104224. [PMID: 38479055 DOI: 10.1016/j.jri.2024.104224] [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: 12/07/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Myelomeningocele (MMC) results from incomplete closure of the neural tube, and has a complex multifactorial etiology, including an inflammatory microenvironment. OBJECTIVE We evaluated the contribution of humoral immune response for development of inflammatory milieu. METHODS Using public repository Gene Expression Omnibus (GEO), we retrieve dataset transcriptome from the amniotic fluid of ten fetuses with myelomeningocele and ten healthy control fetuses to found differential gene expression associated with disturbances and inflammatory signatures in MMC. The identified DEGs were submitted to enrichment, network, and matrix correlation analyses. RESULTS Our initial analysis revealed 90 DEGs in MMC, mainly associated with signaling pathways of inflammation, including the immune modules, humoral immune response and IFN-type I signatures. Protein-protein analysis (PPI) revealed an association with three protein networks; positive regulation of B cell proliferation constituted the largest network. Matrix correlation analyses showed that MMC alters the co-expression of genes related to inflammatory processes that promote microenvironment inflammation. CONCLUSION These results revealed an altered humoral immune response in MMC patients, contributing to an inflammatory profile and providing opportunities for identifying potential biomarkers in myelomeningocele disease.
Collapse
Affiliation(s)
- Rfm Felício
- Birth Defects Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rede de Atenção à Saúde nas Anomalias Congênitas do Sistema Nervoso Central, Instituo Oswaldo Cruz, Fundação Oswaldo Cruz, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Almm Franco
- Birth Defects Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Ciências Biológicas (Genética), Universidade Federal do Rio de Janeiro, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - D Corrêa-Moreira
- Rede de Atenção à Saúde nas Anomalias Congênitas do Sistema Nervoso Central, Instituo Oswaldo Cruz, Fundação Oswaldo Cruz, Brazil; Laboratory of Taxonomy, Biochesmistry and Bioprospecting of Fungi, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - F Martinez de Carvalho
- Birth Defects Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rede de Atenção à Saúde nas Anomalias Congênitas do Sistema Nervoso Central, Instituo Oswaldo Cruz, Fundação Oswaldo Cruz, Brazil; Programa de Pós-Graduação em Ciências Biológicas (Genética), Universidade Federal do Rio de Janeiro, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Acr Guimarães
- Laboratory of Taxonomy, Biochesmistry and Bioprospecting of Fungi, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - F R Vargas
- Birth Defects Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rede de Atenção à Saúde nas Anomalias Congênitas do Sistema Nervoso Central, Instituo Oswaldo Cruz, Fundação Oswaldo Cruz, Brazil; Programa de Pós-Graduação em Ciências Biológicas (Genética), Universidade Federal do Rio de Janeiro, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Departamento de Genética e Biologia Molecular, Universidade Federal do Estado do Rio de Janeiro, Brazil.
| |
Collapse
|
23
|
de Armas LR, Dinh V, Iyer A, Pallikkuth S, Pahwa R, Cotugno N, Rinaldi S, Palma P, Vaz P, Lain MG, Pahwa S. Accelerated CD8 + T cell maturation in infants with perinatal HIV infection. iScience 2024; 27:109720. [PMID: 38706858 PMCID: PMC11068557 DOI: 10.1016/j.isci.2024.109720] [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: 12/06/2023] [Revised: 01/30/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
In perinatal HIV infection, early antiretroviral therapy (ART) initiation is recommended but questions remain regarding infant immune responses to HIV and its impact on immune development. Using single cell transcriptional and phenotypic analysis we evaluated the T cell compartment at pre-ART initiation of infants with perinatally acquired HIV from Maputo, Mozambique (Towards AIDS Remission Approaches cohort). CD8+ T cell maturation subsets exhibited altered distribution in HIV exposed infected (HEI) infants relative to HIV exposed uninfected infants with reduced naive, increased effectors, higher frequencies of activated T cells, and lower frequencies of cells with markers of self-renewal. Additionally, a cluster of CD8+ T cells identified in HEI displayed gene profiles consistent with cytotoxic T lymphocytes and showed evidence for hyper expansion. Longitudinal phenotypic analysis revealed accelerated maturation of CD8+ T cells was maintained in HEI despite viral control. The results point to an HIV-directed immune response that is likely to influence reservoir establishment.
Collapse
Affiliation(s)
- Lesley R. de Armas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vinh Dinh
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Akshay Iyer
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rajendra Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nicola Cotugno
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Stefano Rinaldi
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paula Vaz
- Instituto Nacional de Saúde, Marracuene, Maputo Province, Mozambique
| | | | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
24
|
Vimercati A, Cazzato G, Lospalluti L, Foligno S, Taliento C, Trojanowska KB, Cicinelli E, Bonamonte D, Caliandro D, Vitagliano A, Nicolì P. Dystrophic Epidermolysis Bullosa (DEB): How Can Pregnancy Alter the Course of This Rare Disease? An Updated Literature Review on Obstetrical Management with an Additional Italian Experience. Diseases 2024; 12:104. [PMID: 38785759 PMCID: PMC11120570 DOI: 10.3390/diseases12050104] [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: 04/18/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Epidermolysis Bullosa (EB) is an extremely rare and disabling inherited genetic skin disease with a predisposition to develop bullous lesions on the skin and inner mucous membranes, occurring after mild friction or trauma, or even spontaneously. Within the spectrum of EB forms, dystrophic EB (DEB) represents the most intriguing and challenging in terms of clinical management, especially with regard to pregnancy, due to the highly disabling and life-threatening phenotype. Disappointingly, in the literature little focus has been directed towards pregnancy and childbirth in DEB patients, resulting in a lack of sound evidence and guidance for patients themselves and clinicians. The current study aims to contribute to the DEB literature with an updated summary of the existing evidence regarding the obstetrical and anesthesiological management of this rare disease. Furthermore, this literature review sought to answer the question of whether, and if so, in which way, the pregnancy condition may alter the course of the underlying dermatologic skin disease. Having all this information is indispensable when counseling a patient with DEB who desires a child or is expecting one. Finally, we reported own experience with a pregnant woman with a recessive DEB whom we recently managed, with a favorable outcome.
Collapse
Affiliation(s)
- Antonella Vimercati
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Gerardo Cazzato
- Department of Emergency and Organ Transplantation (DETO), Section of Pathology, University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Lucia Lospalluti
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Dermatology and Venereology, University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Stefania Foligno
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Dermatology and Venereology, University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Cristina Taliento
- Department of Medical Sciences, Obstetrics and Gynecology Unit, University Hospital “Sant’Anna”, 44121 Ferrara, Italy
| | | | - Ettore Cicinelli
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Domenico Bonamonte
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Dermatology and Venereology, University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Dario Caliandro
- Pelvic Floor Center, Department of Gynaecology and Obstetrics, Pia Fondazione “Card. G. Panico”, 73039 Tricase, Italy
| | - Amerigo Vitagliano
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Pierpaolo Nicolì
- Unit of Obstetrics and Gynecology, Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| |
Collapse
|
25
|
Lassoued N, Yero A, Jenabian MA, Soret R, Pilon N. Efficient enzyme-free method to assess the development and maturation of the innate and adaptive immune systems in the mouse colon. Sci Rep 2024; 14:11063. [PMID: 38744932 PMCID: PMC11094196 DOI: 10.1038/s41598-024-61834-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024] Open
Abstract
Researchers who aim to globally analyze the gastrointestinal immune system via flow cytometry have many protocol options to choose from, with specifics generally tied to gut wall layers of interest. To get a clearer idea of the approach we should use on full-thickness colon samples from mice, we first undertook a systematic comparison of three tissue dissociation techniques: two based on enzymatic cocktails and the other one based on manual crushing. Using flow cytometry panels of general markers of lymphoid and myeloid cells, we found that the presence of cell-surface markers and relative cell population frequencies were more stable with the mechanical method. Both enzymatic approaches were associated with a marked decrease of several cell-surface markers. Using mechanical dissociation, we then developed two minimally overlapping panels, consisting of a total of 26 antibodies, for serial profiling of lymphoid and myeloid lineages from the mouse colon in greater detail. Here, we highlight how we accurately delineate these populations by manual gating, as well as the reproducibility of our panels on mouse spleen and whole blood. As a proof-of-principle of the usefulness of our general approach, we also report segment- and life stage-specific patterns of immune cell profiles in the colon. Overall, our data indicate that mechanical dissociation is more suitable and efficient than enzymatic methods for recovering immune cells from all colon layers at once. Additionally, our panels will provide researchers with a relatively simple tool for detailed immune cell profiling in the murine gastrointestinal tract, regardless of life stage or experimental conditions.
Collapse
Affiliation(s)
- Nejia Lassoued
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
| | - Alexis Yero
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
- Human Immuno-Virology Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
- Human Immuno-Virology Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Rodolphe Soret
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada.
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada.
| | - Nicolas Pilon
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada.
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada.
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
26
|
Bakoyan Z, Cao Y, Hansson SR, Karlsson JP, Lodefalk M. Childhood atopic disorders in relation to placental changes-A systematic review and meta-analysis. Pediatr Allergy Immunol 2024; 35:e14141. [PMID: 38773752 DOI: 10.1111/pai.14141] [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: 10/12/2023] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024]
Abstract
Fetal programming may arise from prenatal exposure and increase the risk of diseases later in life, potentially mediated by the placenta. The objective of this systematic review was to summarize and critically evaluate publications describing associations between human placental changes and risk of atopic disorders during childhood. The review adhered to the Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines. The inclusion criteria were original research articles or case reports written in English describing a human placental change in relation to disease occurring in offspring during childhood. The MEDLINE and EMBASE databases were searched for eligible studies. Risk of bias (RoB) was assessed using the ROBINS-I tool. The results were pooled both in a narrative way and by a meta-analysis. Nineteen studies were included (n = 12,997 participants). All studies had an overall serious RoB, and publication bias could not be completely ruled out. However, five studies showed that histological chorioamnionitis in preterm-born children was associated with asthma-related problems (pooled odds ratio = 3.25 (95% confidence interval = 2.22-4.75)). In term-born children, a large placenta (≥750 g) increased the risk of being prescribed anti-asthma medications during the first year of life. Placental histone acetylation, DNA methylation, and gene expression differences were found to be associated with different atopic disorders in term-born children. There is some evidence supporting the idea that the placenta can mediate an increased risk of atopic disorders in children. However, further studies are needed to validate the findings, properly control for confounders, and examine potential mechanisms.
Collapse
Affiliation(s)
- Zaki Bakoyan
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Yang Cao
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Stefan R Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Science Lund, Lund University, Lund, Sweden
| | | | - Maria Lodefalk
- University Health Care Research Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Pediatrics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
27
|
Tisseyre M, Collier M, Beeker N, Kaguelidou F, Treluyer JM, Chouchana L. In Utero Exposure to Antibiotics and Risk of Serious Infections in the First Year of Life. Drug Saf 2024; 47:453-464. [PMID: 38409516 DOI: 10.1007/s40264-024-01401-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/28/2024]
Abstract
INTRODUCTION AND OBJECTIVE Given the high prevalence of antibiotic prescription during pregnancy in France and previous studies suggesting an increased risk of infection in offspring with such exposures, our study aimed to investigate the association between prenatal exposure to systemic antibiotics and serious infections in full-term infants during their first year of life. METHODS We conducted a retrospective population-based cohort study on singleton, full-term liveborn non-immunocompromised infants, using the French National Health Data System (SNDS) between 2012 and 2021. Systemic antibiotic dispensing in ambulatory care settings during pregnancy defined the exposure. Outcomes concerned serious infections (i.e., infections requiring hospitalization) in offspring identified between 3 and 12 months of life, hence excluding infections of maternal origin. Adjusted odds ratios (aORs) were estimated using logistic regression with multivariate models to control for potential confounders. RESULTS Of 2,836,630 infants included, 39.6% were prenatally exposed to systemic antibiotics. Infants prenatally exposed to antibiotics had a higher incidence of serious infections compared with unexposed infants {aOR 1.12 [95% confidence interval (95% CI) 1.11-1.13]}. Similar associations were observed according to the timing of exposure during pregnancy, antibiotic class, and site of infections. The strongest association was observed when infants were prenatally exposed to three or more antibiotic courses during pregnancy [aOR 1.21 (95% CI 1.19-1.24)]. Limitations include residual confounders, such as genetic susceptibility to infections and the role of the underlying pathogen agent. CONCLUSION Prenatal exposure to systemic antibiotics is very common and is associated with a weak yet significant associations with subsequent serious infectious events during the first year of life. While our study revealed associations, it is important to note that causation cannot be established, given the acknowledged limitations, including potential confounding by indication.
Collapse
Affiliation(s)
- Mylène Tisseyre
- Centre Régional de Pharmacovigilance, Service de Pharmacologie périnatale, pédiatrique et adulte, Hopital Cochin, Assistance Publique-Hopitaux de Paris (AP-HP), 27, rue du Faubourg Saint Jacques, 75014, Paris, France.
- EA7323, Evaluation thérapeutique et pharmacologie périnatale et pédiatrique, Université Paris Cité, Paris, France.
| | - Mathis Collier
- EA7323, Evaluation thérapeutique et pharmacologie périnatale et pédiatrique, Université Paris Cité, Paris, France
- Unité de Recherche Clinique, Hopital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Nathanaël Beeker
- EA7323, Evaluation thérapeutique et pharmacologie périnatale et pédiatrique, Université Paris Cité, Paris, France
- Unité de Recherche Clinique, Hopital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Florentia Kaguelidou
- EA7323, Evaluation thérapeutique et pharmacologie périnatale et pédiatrique, Université Paris Cité, Paris, France
- Centre d'Investigations Cliniques, INSERM CIC1426, Hôpital Robert Debré, APHP.Nord, Paris, France
| | - Jean-Marc Treluyer
- Centre Régional de Pharmacovigilance, Service de Pharmacologie périnatale, pédiatrique et adulte, Hopital Cochin, Assistance Publique-Hopitaux de Paris (AP-HP), 27, rue du Faubourg Saint Jacques, 75014, Paris, France
- EA7323, Evaluation thérapeutique et pharmacologie périnatale et pédiatrique, Université Paris Cité, Paris, France
- Unité de Recherche Clinique, Hopital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Laurent Chouchana
- Centre Régional de Pharmacovigilance, Service de Pharmacologie périnatale, pédiatrique et adulte, Hopital Cochin, Assistance Publique-Hopitaux de Paris (AP-HP), 27, rue du Faubourg Saint Jacques, 75014, Paris, France
- EA7323, Evaluation thérapeutique et pharmacologie périnatale et pédiatrique, Université Paris Cité, Paris, France
| |
Collapse
|
28
|
DaMata JP, Zelkoski AE, Nhan PB, Ennis KHE, Kim JS, Lu Z, Malloy AMW. Dissociation protocols influence the phenotypes of lymphocyte and myeloid cell populations isolated from the neonatal lymph node. Front Immunol 2024; 15:1368118. [PMID: 38756770 PMCID: PMC11097666 DOI: 10.3389/fimmu.2024.1368118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Frequencies and phenotypes of immune cells differ between neonates and adults in association with age-specific immune responses. Lymph nodes (LN) are critical tissue sites to quantify and define these differences. Advances in flow cytometry have enabled more multifaceted measurements of complex immune responses. Tissue processing can affect the immune cells under investigation that influence key findings. To understand the impact on immune cells in the LN after processing for single-cell suspension, we compared three dissociation protocols: enzymatic digestion, mechanical dissociation with DNase I treatment, and mechanical dissociation with density gradient separation. We analyzed cell yields, viability, phenotypic and maturation markers of immune cells from the lung-draining LN of neonatal and adult mice two days after intranasal respiratory syncytial virus (RSV) infection. While viability was consistent across age groups, the protocols influenced the yield of subsets defined by important phenotypic and activation markers. Moreover, enzymatic digestion did not show higher overall yields of conventional dendritic cells and macrophages from the LN. Together, our findings show that the three dissociation protocols have similar impacts on the number and viability of cells isolated from the neonatal and adult LN. However, enzymatic digestion impacts the mean fluorescence intensity of key lineage and activation markers that may influence experimental findings.
Collapse
Affiliation(s)
- Jarina P. DaMata
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Amanda E. Zelkoski
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| | - Paula B. Nhan
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Katherine H. E. Ennis
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Ji Sung Kim
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Zhongyan Lu
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Allison M. W. Malloy
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| |
Collapse
|
29
|
Wei J, Zhang L, Xu H, Luo Q. Preterm birth, a consequence of immune deviation mediated hyperinflammation. Heliyon 2024; 10:e28483. [PMID: 38689990 PMCID: PMC11059518 DOI: 10.1016/j.heliyon.2024.e28483] [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: 12/21/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 05/02/2024] Open
Abstract
Preterm birth represents a multifaceted syndrome with intricacies still present in our comprehension of its etiology. In the context of a semi-allograft, the prosperity from implantation to pregnancy to delivery hinges on the establishment of a favorable maternal-fetal immune microenvironment and a successful trilogy of immune activation, immune tolerance and then immune activation transitions. The occurrence of spontaneous preterm birth could be related to abnormalities within the immune trilogy, stemming from deviation in maternal and fetal immunity. These immune deviations, characterized by insufficient immune tolerance and early immune activation, ultimately culminated in an unsustainable pregnancy. In this review, we accentuated the role of both innate and adaptive immune reason in promoting spontaneous preterm birth, reviewed the risk of preterm birth from vaginal microbiome mediated by immune changes and the potential of vaginal microbiomes and metabolites as a new predictive marker, and discuss the changes in the role of progesterone and its interaction with immune cells in a preterm birth population. Our objective was to contribute to the growing body of knowledge in the field, shedding light on the immunologic reason of spontaneous preterm birth and effective biomarkers for early prediction, providing a roadmap for forthcoming investigations.
Collapse
Affiliation(s)
- Juan Wei
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| | - LiYuan Zhang
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| | - Heng Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Qiong Luo
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| |
Collapse
|
30
|
Ho SJ, Chaput D, Sinkey RG, Garces AH, New EP, Okuka M, Sang P, Arlier S, Semerci N, Steffensen TS, Rutherford TJ, Alsina AE, Cai J, Anderson ML, Magness RR, Uversky VN, Cummings DAT, Tsibris JCM. Proteomic studies of VEGFR2 in human placentas reveal protein associations with preeclampsia, diabetes, gravidity, and labor. Cell Commun Signal 2024; 22:221. [PMID: 38594674 PMCID: PMC11003095 DOI: 10.1186/s12964-024-01567-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024] Open
Abstract
VEGFR2 (Vascular endothelial growth factor receptor 2) is a central regulator of placental angiogenesis. The study of the VEGFR2 proteome of chorionic villi at term revealed its partners MDMX (Double minute 4 protein) and PICALM (Phosphatidylinositol-binding clathrin assembly protein). Subsequently, the oxytocin receptor (OT-R) and vasopressin V1aR receptor were detected in MDMX and PICALM immunoprecipitations. Immunogold electron microscopy showed VEGFR2 on endothelial cell (EC) nuclei, mitochondria, and Hofbauer cells (HC), tissue-resident macrophages of the placenta. MDMX, PICALM, and V1aR were located on EC plasma membranes, nuclei, and HC nuclei. Unexpectedly, PICALM and OT-R were detected on EC projections into the fetal lumen and OT-R on 20-150 nm clusters therein, prompting the hypothesis that placental exosomes transport OT-R to the fetus and across the blood-brain barrier. Insights on gestational complications were gained by univariable and multivariable regression analyses associating preeclampsia with lower MDMX protein levels in membrane extracts of chorionic villi, and lower MDMX, PICALM, OT-R, and V1aR with spontaneous vaginal deliveries compared to cesarean deliveries before the onset of labor. We found select associations between higher MDMX, PICALM, OT-R protein levels and either gravidity, diabetes, BMI, maternal age, or neonatal weight, and correlations only between PICALM-OT-R (p < 2.7 × 10-8), PICALM-V1aR (p < 0.006), and OT-R-V1aR (p < 0.001). These results offer for exploration new partnerships in metabolic networks, tissue-resident immunity, and labor, notably for HC that predominantly express MDMX.
Collapse
Grants
- Department of Obstetrics and Gynecology, University of South Florida
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida
- Lisa Muma Weitz Microscopy Laboratory, University of South Florida
- Department of Chemistry, University of South Florida
- Tampa General Hospital, Tampa, Florida
- Teasley Foundation
- Department of Molecular Medicine, University of South Florida
- Department of Biology, University of Florida
- Emerging Pathogens Institute, University of Florida
Collapse
Affiliation(s)
- Shannon J Ho
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Rachel G Sinkey
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Amanda H Garces
- Lisa Muma Weitz Microscopy Laboratory, University of South Florida, Tampa, FL, USA
| | - Erika P New
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Maja Okuka
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Peng Sang
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Sefa Arlier
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Nihan Semerci
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | | | - Thomas J Rutherford
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
- Cancer Center, Tampa General Hospital, Tampa, FL, USA
| | - Angel E Alsina
- Transplant Surgery Center, Tampa General Hospital, Tampa, FL, USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Matthew L Anderson
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
- Cancer Center, Tampa General Hospital, Tampa, FL, USA
| | - Ronald R Magness
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Derek A T Cummings
- Department of Biology and Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - John C M Tsibris
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA.
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
31
|
Arakawa A, Tao K, Kohno T, Ogawa C. Cross-individual cancer transmission to children during the gestational and perinatal periods. Cancer Sci 2024; 115:1039-1047. [PMID: 38369705 PMCID: PMC11006992 DOI: 10.1111/cas.16102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/13/2024] [Accepted: 01/25/2024] [Indexed: 02/20/2024] Open
Abstract
Cancer transmission may rarely occur between individuals. Besides through allogenic transplantation, cancer transmission via the hemochorial placenta, which is permissive for cell traffic, has been described in a few reports. Three etiologies of transplacental cancer transmission include (1) maternofetal transmission of maternal cancer cells, (2) transmission of gestational choriocarcinoma to the fetus, and (3) transfer of preleukemic cells from one monozygotic twin to the other. Additionally, we recently reported two pediatric cases of lung tumors in which the lung-only distribution of tumors and genomic profiling of both the child's and mother's tumor samples suggested the airway/transbronchial transmission of maternal cervical cancer cells to the child by aspiration at birth. The immune system coordinates the hemostatic balance between effector and regulatory immunity, especially during fetal development. The immunoregulatory properties are shared in both physiological pregnancy-related and pathological cancer-related conditions. Mechanistically, the survival and colonization of transmitted cancer cells within a child are likely attributed to a combination of the child's immune tolerance and the cancer's immune escape. In this review, we summarize the current understanding of gestational/perinatal cancer transmission and discuss the possible mechanism-based immunotherapy for this rare form of pediatric cancer.
Collapse
Affiliation(s)
- Ayumu Arakawa
- Department of Pediatric OncologyNational Cancer Center HospitalTokyoJapan
| | - Kayoko Tao
- Department of Pediatric OncologyNational Cancer Center HospitalTokyoJapan
| | - Takashi Kohno
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
| | - Chitose Ogawa
- Department of Pediatric OncologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
32
|
Hinshaw DC, Patel M, Shevde LA. A Metabolic Axis of Immune Intractability. Cancer Immunol Res 2024; 12:282-286. [PMID: 38126910 PMCID: PMC10936744 DOI: 10.1158/2326-6066.cir-23-0433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/28/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Immune cells in the tumor niche robustly influence disease progression. Remarkably, in cancer, developmental pathways are reenacted. Many parallels between immune regulation of embryonic development and immune regulation of tumor progression can be drawn, with evidence clearly supporting an immune-suppressive microenvironment in both situations. In these ecosystems, metabolic and bioenergetic circuits guide and regulate immune cell differentiation, plasticity, and functional properties of suppressive and inflammatory immune subsets. As such, there is an emerging pattern of intersection across the dynamic process of ontogeny and the ever-evolving tumor neighborhood. In this article, we focus on the convergence of immune programming during ontogeny and in the tumor microenvironment. Exemplifying dysregulation of Hedgehog (Hh) activity, a key player during ontogeny, we highlight a critical convergence of these fields and the metabolic axis of the nutrient sensing hexosamine biosynthetic pathway (HBP) that integrates glucose, glutamine, amino acids, acetyl CoA, and uridine-5'-triphosphate (UTP), culminating in the synthesis of UDP-GlcNAc, a metabolite that functions as a metabolic and bioenergetic sensor. We discuss an emerging pattern of immune regulation, orchestrated by O-GlcNAcylation of key transcriptional regulators, spurring suppressive activity of dysfunctional immune cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Dominique C. Hinshaw
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Meet Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A. Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Senior author
| |
Collapse
|
33
|
Quincer EM, Cranmer LM, Kamidani S. Prenatal Maternal Immunization for Infant Protection: A Review of the Vaccines Recommended, Infant Immunity and Future Research Directions. Pathogens 2024; 13:200. [PMID: 38535543 PMCID: PMC10975994 DOI: 10.3390/pathogens13030200] [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: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 04/01/2024] Open
Abstract
Prenatal maternal immunization is an effective tool to protect mothers and infants from poor health outcomes due to infectious diseases. We provide an overview of the rationale for the use of prenatal vaccines, discuss the immunologic environment of the maternal-fetal interface including the impact of maternal vaccines prenatally and subsequently on the infant's immune response, and review vaccines currently recommended in pregnancy and landscape for the future of maternal vaccination. This review aims to provide an understanding of the recent history and progress made in the field and highlight the importance of continued research and development into new vaccines for pregnant populations.
Collapse
Affiliation(s)
- Elizabeth M. Quincer
- Immunization Safety Office, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Lisa M. Cranmer
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Satoshi Kamidani
- Immunization Safety Office, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
34
|
Liu J, Joseph S, Manohar K, Lee J, Brokaw JP, Shelley WC, Markel TA. Role of innate T cells in necrotizing enterocolitis. Front Immunol 2024; 15:1357483. [PMID: 38390341 PMCID: PMC10881895 DOI: 10.3389/fimmu.2024.1357483] [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: 12/18/2023] [Accepted: 01/16/2024] [Indexed: 02/24/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a destructive gastrointestinal disease primarily affecting preterm babies. Despite advancements in neonatal care, NEC remains a significant cause of morbidity and mortality in neonatal intensive care units worldwide and the etiology of NEC is still unclear. Risk factors for NEC include prematurity, very low birth weight, feeding with formula, intestinal dysbiosis and bacterial infection. A review of the literature would suggest that supplementation of prebiotics and probiotics prevents NEC by altering the immune responses. Innate T cells, a highly conserved subpopulation of T cells that responds quickly to stimulation, develops differently from conventional T cells in neonates. This review aims to provide a succinct overview of innate T cells in neonates, encompassing their phenotypic characteristics, functional roles, likely involvement in the pathogenesis of NEC, and potential therapeutic implications.
Collapse
Affiliation(s)
- Jianyun Liu
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sharon Joseph
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Krishna Manohar
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jasmine Lee
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - John P. Brokaw
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - W. Christopher Shelley
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
| | - Troy A. Markel
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
| |
Collapse
|
35
|
Ohta R, Fujimori T, Sano C. Eosinophilic Granulomatosis With Polyangiitis Gradually Worsening With Consecutive Pregnancies: A Case Report. Cureus 2024; 16:e54832. [PMID: 38529439 PMCID: PMC10963072 DOI: 10.7759/cureus.54832] [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] [Accepted: 02/24/2024] [Indexed: 03/27/2024] Open
Abstract
This case report elucidates the diagnosis and management of eosinophilic granulomatosis with polyangiitis (EGPA), a form of systemic vasculitis, in a 32-year-old female presenting with progressive dermal, respiratory, and gastrointestinal symptoms following multiple pregnancies. The patient's history of allergic reactions and pregnancies suggested a gradual progression of EGPA, a condition rarely associated with pregnancy. Initial symptoms were misinterpreted as allergic reactions and acute gastroenteritis, delaying the correct diagnosis. Laboratory findings included eosinophilia and elevated immunoglobulin E, while further investigations ruled out other differential diagnoses, such as chronic eosinophilic leukemia. A clinical diagnosis of EGPA was made based on symptom progression, eosinophilia, and mononeuritis multiplex, absent typical granulomatous changes in the skin biopsy. Treatment with high-dose prednisolone and rituximab halted disease progression and improved symptoms, highlighting the critical importance of prompt diagnosis and treatment in preventing irreversible complications. This case emphasizes the need for general physicians to consider pregnancy as a potential trigger for autoimmune diseases like EGPA, especially in patients presenting with multi-symptom allergic reactions and high inflammatory markers.
Collapse
Affiliation(s)
| | | | - Chiaki Sano
- Community Medicine Management, Shimane University Faculty of Medicine, Izumo, JPN
| |
Collapse
|
36
|
Koren O, Konnikova L, Brodin P, Mysorekar IU, Collado MC. The maternal gut microbiome in pregnancy: implications for the developing immune system. Nat Rev Gastroenterol Hepatol 2024; 21:35-45. [PMID: 38097774 DOI: 10.1038/s41575-023-00864-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 01/04/2024]
Abstract
The gut microbiome has important roles in host metabolism and immunity, and microbial dysbiosis affects human physiology and health. Maternal immunity and microbial metabolites during pregnancy, microbial transfer during birth, and transfer of immune factors, microorganisms and metabolites via breastfeeding provide critical sources of early-life microbial and immune training, with important consequences for human health. Only a few studies have directly examined the interactions between the gut microbiome and the immune system during pregnancy, and the subsequent effect on offspring development. In this Review, we aim to describe how the maternal microbiome shapes overall pregnancy-associated maternal, fetal and early neonatal immune systems, focusing on the existing evidence and highlighting current gaps to promote further research.
Collapse
Affiliation(s)
- Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Liza Konnikova
- Department of Paediatrics and Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Petter Brodin
- Department of Immunology and Inflammation, Imperial College London, London, UK
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
37
|
Waddington SN, Peranteau WH, Rahim AA, Boyle AK, Kurian MA, Gissen P, Chan JKY, David AL. Fetal gene therapy. J Inherit Metab Dis 2024; 47:192-210. [PMID: 37470194 PMCID: PMC10799196 DOI: 10.1002/jimd.12659] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Fetal gene therapy was first proposed toward the end of the 1990s when the field of gene therapy was, to quote the Gartner hype cycle, at its "peak of inflated expectations." Gene therapy was still an immature field but over the ensuing decade, it matured and is now a clinical and market reality. The trajectory of treatment for several genetic diseases is toward earlier intervention. The ability, capacity, and the will to diagnose genetic disease early-in utero-improves day by day. A confluence of clinical trials now signposts a trajectory toward fetal gene therapy. In this review, we recount the history of fetal gene therapy in the context of the broader field, discuss advances in fetal surgery and diagnosis, and explore the full ambit of preclinical gene therapy for inherited metabolic disease.
Collapse
Affiliation(s)
- Simon N Waddington
- EGA Institute for Women's Health, University College London, London, UK
- Faculty of Health Sciences, Wits/SAMRC Antiviral Gene Therapy Research Unit, Johannesburg, South Africa
| | - William H Peranteau
- The Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, London, UK
| | - Ashley K Boyle
- EGA Institute for Women's Health, University College London, London, UK
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, UK
- Department of Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Paul Gissen
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London, UK
| | - Jerry K Y Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
- Academic Clinical Program in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore, Singapore
- Experimental Fetal Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anna L David
- EGA Institute for Women's Health, University College London, London, UK
| |
Collapse
|
38
|
Gostomczyk K, Borowczak J, Siekielska-Domanowska M, Szczerbowski K, Maniewski M, Dubiel M, Szylberg Ł, Bodnar M. Mechanisms of SARS-CoV-2 Placental Transmission. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0001. [PMID: 38299561 DOI: 10.2478/aite-2024-0001] [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: 06/30/2023] [Accepted: 09/07/2023] [Indexed: 02/02/2024]
Abstract
The widespread occurrence of SARS-CoV-2 infections and the diverse range of symptoms have placed significant strain on healthcare systems worldwide. Pregnancy has also been affected by COVID-19, with an increased risk of complications and unfavorable outcomes for expectant mothers. Multiple studies indicate that SARS-CoV-2 can infiltrate the placenta, breach its protective barrier, and infect the fetus. Although the precise mechanisms of intrauterine transmission remain unclear, factors such as perinatal infection, macrophages, sexual intercourse, and the virus' interaction with host angiotensin-converting enzyme 2 (ACE2) and neuropilin-1 (NRP-1) proteins appear to play a role in this process. The integrity of the placental barrier fluctuates throughout pregnancy and appears to influence the likelihood of fetal transmission. The expression of placental cell receptors, like ACE2, changes during pregnancy and in response to placental damage. However, due to the consistent presence of others, such as NRP-1, SARS-CoV-2 may potentially enter the fetus at different stages of pregnancy. NRP-1 is also found in macrophages, implicating maternal macrophages and Hofbauer cells as potential routes for viral transmission. Our current understanding of SARS-CoV-2's vertical transmission pathways remains limited. Some researchers question the ACE2-associated transmission model due to the relatively low expression of ACE2 in the placenta. Existing studies investigating perinatal transmission and the impact of sexual intercourse have either involved small sample sizes or lacked statistical significance. This review aims to explore the current state of knowledge regarding the potential mechanisms of COVID-19 vertical transmission, identifying areas where further research is needed to fill the gaps in our understanding.
Collapse
Affiliation(s)
- Karol Gostomczyk
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
- Department of Tumor Pathology and Pathomorphology, Oncology Centre - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
| | - Jędrzej Borowczak
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Marta Siekielska-Domanowska
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Krzysztof Szczerbowski
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Mateusz Maniewski
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Mariusz Dubiel
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Łukasz Szylberg
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
- Department of Tumor Pathology and Pathomorphology, Oncology Centre - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
- Chair of Pathology, Dr. Jan Biziel Memorial University Hospital No. 2, Bydgoszcz, Poland
| | - Magdalena Bodnar
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
- Chair of Pathology, Dr. Jan Biziel Memorial University Hospital No. 2, Bydgoszcz, Poland
| |
Collapse
|
39
|
Prigge AD, Coates BM. Can Airway Monocytes from Premature Infants Teach Us about Macrophage Differentiation in the Lungs? Am J Respir Cell Mol Biol 2023; 69:612-613. [PMID: 37702543 DOI: 10.1165/rcmb.2023-0305ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Affiliation(s)
- Andrew D Prigge
- Department of Pediatrics Northwestern University Chicago, Illinois
- Ann & Robert H. Lurie Children's Hospital of Chicago Chicago, Illinois
| | - Bria M Coates
- Department of Pediatrics Northwestern University Chicago, Illinois
- Ann & Robert H. Lurie Children's Hospital of Chicago Chicago, Illinois
| |
Collapse
|
40
|
Donald K, Finlay BB. Early-life interactions between the microbiota and immune system: impact on immune system development and atopic disease. Nat Rev Immunol 2023; 23:735-748. [PMID: 37138015 DOI: 10.1038/s41577-023-00874-w] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2023] [Indexed: 05/05/2023]
Abstract
Prenatal and early postnatal life represent key periods of immune system development. In addition to genetics and host biology, environment has a large and irreversible role in the immune maturation and health of an infant. One key player in this process is the gut microbiota, a diverse community of microorganisms that colonizes the human intestine. The diet, environment and medical interventions experienced by an infant determine the establishment and progression of the intestinal microbiota, which interacts with and trains the developing immune system. Several chronic immune-mediated diseases have been linked to an altered gut microbiota during early infancy. The recent rise in allergic disease incidence has been explained by the 'hygiene hypothesis', which states that societal changes in developed countries have led to reduced early-life microbial exposures, negatively impacting immunity. Although human cohort studies across the globe have established a correlation between early-life microbiota composition and atopy, mechanistic links and specific host-microorganism interactions are still being uncovered. Here, we detail the progression of immune system and microbiota maturation in early life, highlight the mechanistic links between microbes and the immune system, and summarize the role of early-life host-microorganism interactions in allergic disease development.
Collapse
Affiliation(s)
- Katherine Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
41
|
Grama A, Mititelu A, Sîrbe C, Benţa G, Pop TL. Immune-mediated cholangiopathies in children: the need to better understand the pathophysiology for finding the future possible treatment targets. Front Immunol 2023; 14:1206025. [PMID: 37928553 PMCID: PMC10623351 DOI: 10.3389/fimmu.2023.1206025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023] Open
Abstract
Cholangiopathies are defined as focal or extensive damage of the bile ducts. According to the pathogenetic mechanism, it may be immune-mediated or due to genetic, infectious, toxic, vascular, and obstructive causes. Their chronic evolution is characterized by inflammation, obstruction of bile flow, cholangiocyte proliferation, and progression toward fibrosis and cirrhosis. Immune-mediated cholangiopathies comprise primary sclerosing cholangitis (PSC), autoimmune cholangitis and IgG4-associated cholangitis in adults and biliary atresia (BA), neonatal sclerosing cholangitis (NSC) in children. The main purpose of this narrative review was to highlight the similarities and differences among immune-mediated cholangiopathies, especially those frequent in children in which cholangiocyte senescence plays a key role (BA, NSC, and PSC). These three entities have many similarities in terms of clinical and histopathological manifestations, and the distinction between them can be hard to achieve. In BA, bile duct destruction occurs due to aggression of the biliary cells due to viral infections or toxins during the intrauterine period or immediately after birth. The consequence is the activation of the immune system leading to severe inflammation and fibrosis of the extrahepatic biliary tract, lumen stenosis, and impairment of the biliary flow. PSC is characterized by inflammation and fibrosis of intra- and extrahepatic bile ducts, leading to secondary biliary cirrhosis. It is a multifactorial disease that occurs because of genetic predisposition [human leukocyte antigen (HLA) and non-HLA haplotypes], autoimmunity (cellular immune response, autoantibodies, association with inflammatory bowel disease), environmental factors (infections or toxic bile), and host factors (intestinal microbiota). NSC seems to be a distinct subgroup of childhood PSC that appears due to the interaction between genetic predisposition (HLA B8 and DR3) and the disruption of the immune system, validated by elevated IgG levels or specific antibodies [antinuclear antibody (ANA), anti-smooth muscle antibody (ASMA)]. Currently, the exact mechanism of immune cholangiopathy is not fully understood, and further data are required to identify individuals at high risk of developing these conditions. A better understanding of the immune mechanisms and pathophysiology of BA, NSC, and PSC will open new perspectives for future treatments and better methods of preventing severe evolution.
Collapse
Affiliation(s)
- Alina Grama
- 2Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- 2Pediatric Clinic and Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
| | - Alexandra Mititelu
- 2Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- 2Pediatric Clinic and Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
| | - Claudia Sîrbe
- 2Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- 2Pediatric Clinic and Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
| | - Gabriel Benţa
- 2Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- 2Pediatric Clinic and Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
| | - Tudor Lucian Pop
- 2Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- 2Pediatric Clinic and Center of Expertise in Pediatric Liver Rare Disorders, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
| |
Collapse
|
42
|
Dhariwala MO, DeRogatis AM, Okoro JN, Weckel A, Tran VM, Habrylo I, Ojewumi OT, Tammen AE, Leech JM, Merana GR, Carale RO, Barrere-Cain R, Hiam-Galvez KJ, Spitzer MH, Scharschmidt TC. Commensal myeloid crosstalk in neonatal skin regulates long-term cutaneous type 17 inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560039. [PMID: 37873143 PMCID: PMC10592812 DOI: 10.1101/2023.09.29.560039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Early life microbe-immune interactions at barrier surfaces have lasting impacts on the trajectory towards health versus disease. Monocytes, macrophages and dendritic cells are primary sentinels in barrier tissues, yet the salient contributions of commensal-myeloid crosstalk during tissue development remain poorly understood. Here, we identify that commensal microbes facilitate accumulation of a population of monocytes in neonatal skin. Transient postnatal depletion of these monocytes resulted in heightened IL-17A production by skin T cells, which was particularly sustained among CD4+ T cells into adulthood and sufficient to exacerbate inflammatory skin pathologies. Neonatal skin monocytes were enriched in expression of negative regulators of the IL-1 pathway. Functional in vivo experiments confirmed a key role for excessive IL-1R1 signaling in T cells as contributing to the dysregulated type 17 response in neonatal monocyte-depleted mice. Thus, a commensal-driven wave of monocytes into neonatal skin critically facilitates long-term immune homeostasis in this prominent barrier tissue.
Collapse
Affiliation(s)
- Miqdad O. Dhariwala
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Andrea M. DeRogatis
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Joy N. Okoro
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
| | - Antonin Weckel
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Victoria M. Tran
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
| | - Irek Habrylo
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
| | | | - Allison E. Tammen
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - John M. Leech
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Geil R. Merana
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
| | - Ricardo O. Carale
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Rio Barrere-Cain
- Department of Dermatology, University of California San Francisco; San Francisco, CA USA
| | - Kamir J. Hiam-Galvez
- Biomedical Sciences Program, University of California San Francisco; San Francisco, CA USA
- Department of Otolaryngology-Head and Neck Surgery, Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California San Francisco; San Francisco, CA USA
| | - Matthew H. Spitzer
- Department of Otolaryngology-Head and Neck Surgery, Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California San Francisco; San Francisco, CA USA
| | | |
Collapse
|
43
|
Notarbartolo V, Carta M, Accomando S, Giuffrè M. The First 1000 Days of Life: How Changes in the Microbiota Can Influence Food Allergy Onset in Children. Nutrients 2023; 15:4014. [PMID: 37764797 PMCID: PMC10534753 DOI: 10.3390/nu15184014] [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: 08/04/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Allergic disease, including food allergies (FA)s, has been identified as a major global disease. The first 1000 days of life can be a "window of opportunity" or a "window of susceptibility", during which several factors can predispose children to FA development. Changes in the composition of the gut microbiota from pregnancy to infancy may play a pivotal role in this regard: some bacterial genera, such as Lactobacillus and Bifidobacterium, seem to be protective against FA development. On the contrary, Clostridium and Staphylococcus appear to be unprotective. METHODS We conducted research on the most recent literature (2013-2023) using the PubMed and Scopus databases. We included original papers, clinical trials, meta-analyses, and reviews in English. Case reports, series, and letters were excluded. RESULTS During pregnancy, the maternal diet can play a fundamental role in influencing the gut microbiota composition of newborns. After birth, human milk can promote the development of protective microbial species via human milk oligosaccharides (HMOs), which play a prebiotic role. Moreover, complementary feeding can modify the gut microbiota's composition. CONCLUSIONS The first two years of life are a critical period, during which several factors can increase the risk of FA development in genetically predisposed children.
Collapse
Affiliation(s)
- Veronica Notarbartolo
- Neonatal Intensive Care Unit with Neonatology, “G.F. Ingrassia” Hospital Unit, ASP 6, 90131 Palermo, Italy;
| | - Maurizio Carta
- Neonatology and Neonatal Intensive Care Unit, University Hospital Policlinic “Paolo Giaccone”, 90127 Palermo, Italy;
| | - Salvatore Accomando
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
| | - Mario Giuffrè
- Neonatology and Neonatal Intensive Care Unit, University Hospital Policlinic “Paolo Giaccone”, 90127 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
44
|
Di Lollo V, Canciello A, Peserico A, Orsini M, Russo V, Cerveró-Varona A, Dufrusine B, El Khatib M, Curini V, Mauro A, Berardinelli P, Tournier C, Ancora M, Cammà C, Dainese E, Mincarelli LF, Barboni B. Unveiling the immunomodulatory shift: Epithelial-mesenchymal transition Alters immune mechanisms of amniotic epithelial cells. iScience 2023; 26:107582. [PMID: 37680464 PMCID: PMC10481295 DOI: 10.1016/j.isci.2023.107582] [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: 10/07/2021] [Revised: 06/01/2023] [Accepted: 08/04/2023] [Indexed: 09/09/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) changes cell phenotype by affecting immune properties of amniotic epithelial cells (AECs). The present study shows how the response to lipopolysaccharide of cells collected pre- (eAECs) and post-EMT (mAECs) induces changes in their transcriptomics profile. In fact, eAECs mainly upregulate genes involved in antigen-presenting response, whereas mAECs over-express soluble inflammatory mediator transcripts. Consistently, network analysis identifies CIITA and Nrf2 as main drivers of eAECs and mAECs immune response, respectively. As a consequence, the depletion of CIITA and Nrf2 impairs the ability of eAECs and mAECs to inhibit lymphocyte proliferation or macrophage-dependent IL-6 release, thus confirming their involvement in regulating immune response. Deciphering the mechanisms controlling the immune function of AECs pre- and post-EMT represents a step forward in understanding key physiological events wherein these cells are involved (pregnancy and labor). Moreover, controlling the immunomodulatory properties of eAECs and mAECs may be essential in developing potential strategies for regenerative medicine applications.
Collapse
Affiliation(s)
- Valeria Di Lollo
- National Reference Center for Whole Genome Sequencing of Microbial Pathogens: Database and Bioinformatic Analysis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Campo Boario, 64100 Teramo, Italy
| | - Angelo Canciello
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Alessia Peserico
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Massimiliano Orsini
- National Reference Center for Whole Genome Sequencing of Microbial Pathogens: Database and Bioinformatic Analysis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Campo Boario, 64100 Teramo, Italy
- Istituto Zooprofilattico Sperimentale delle Venezie, Department of Microbiology, Viale dell’Università 10, 35020 Legnaro (PD), Italy
| | - Valentina Russo
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Adrián Cerveró-Varona
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Beatrice Dufrusine
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Mohammad El Khatib
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Valentina Curini
- National Reference Center for Whole Genome Sequencing of Microbial Pathogens: Database and Bioinformatic Analysis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Campo Boario, 64100 Teramo, Italy
| | - Annunziata Mauro
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Paolo Berardinelli
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Massimo Ancora
- National Reference Center for Whole Genome Sequencing of Microbial Pathogens: Database and Bioinformatic Analysis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Campo Boario, 64100 Teramo, Italy
| | - Cesare Cammà
- National Reference Center for Whole Genome Sequencing of Microbial Pathogens: Database and Bioinformatic Analysis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Campo Boario, 64100 Teramo, Italy
| | - Enrico Dainese
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Luana Fiorella Mincarelli
- National Reference Center for Whole Genome Sequencing of Microbial Pathogens: Database and Bioinformatic Analysis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Campo Boario, 64100 Teramo, Italy
| | - Barbara Barboni
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| |
Collapse
|
45
|
Sinkora M, Toman M, Stepanova K, Stepanova H, Leva L, Sinkorova J, Moutelikova R, Salat J, Srutkova D, Schwarzer M, Sinkora S, Skalnikova HK, Nechvatalova K, Hudcovic T, Hermanova P, Pfeiferova S, Kratochvilova M, Kavanova L, Dusankova B, Sinkora MJ. The mechanism of immune dysregulation caused by porcine reproductive and respiratory syndrome virus (PRRSV). Microbes Infect 2023; 25:105146. [PMID: 37142116 DOI: 10.1016/j.micinf.2023.105146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
PRRSV is capable of evading the effective immune response, thus persisting in piglets and throughout the swine herd. We show here that PRRSV invades the thymus and causes depletion of T-cell precursors and alteration of the TCR repertoire. Developing thymocytes are affected during negative selection when they transit from the triple-negative to triple-positive stages at the corticomedullary junction just before entering the medulla. The restriction of repertoire diversification occurs in both helper and cytotoxic αβ-T cells. As a result, critical viral epitopes are tolerated, and infection becomes chronic. However, not all viral epitopes are tolerated. Infected piglets develop antibodies capable of recognizing PRRSV, but these are not virus neutralizing. Further analysis showed that the lack of an effective immune response against the critical viral structures results in the absence of a germinal center response, overactivation of T and B cells in the periphery, robust production of useless antibodies of all isotypes, and the inability to eliminate the virus. Overall, the results show how a respiratory virus that primarily infects and destroys myelomonocytic cells has evolved strategies to disrupt the immune system. These mechanisms may be a prototype for how other viruses can similarly modulate the host immune system.
Collapse
Affiliation(s)
- Marek Sinkora
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic.
| | - Miroslav Toman
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Katerina Stepanova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Hana Stepanova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Lenka Leva
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Jana Sinkorova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Romana Moutelikova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Jiri Salat
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Simon Sinkora
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Helena Kupcova Skalnikova
- Laboratory of Applied Proteome Analyses and Research Center PIGMOD, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Libechov, Czech Republic
| | - Katerina Nechvatalova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Tomas Hudcovic
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Petra Hermanova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Sarka Pfeiferova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Mirka Kratochvilova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Lenka Kavanova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Blanka Dusankova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Marek Jr Sinkora
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czech Republic
| |
Collapse
|
46
|
Li X, Xiao Z, Li C, Chen Q, Jia L. Maternal dietary patterns during pregnancy and the risk of infantile eczema during the first year of life: a cohort study in northeast China. BMC Public Health 2023; 23:1641. [PMID: 37641073 PMCID: PMC10463679 DOI: 10.1186/s12889-023-16577-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND There are few studies on the relationship between diet during pregnancy and infantile eczema and the conclusions are inconsistent. The aim of the present study was to explore the impact of dietary patterns during pregnancy on infantile eczema. METHODS A total of 495 mother-child pairs from a prospective cohort in Shenyang, China was recruited. Information on maternal dietary intake during pregnancy was assessed with a validated self-administered food frequency questionnaire. The data of infantile eczema was assessed using a structured questionnaire. Factor analysis to derive dietary patterns. The relationship between the dietary pattern and infantile eczema was examined by the logistic regression analysis. RESULTS The cumulative incidence of eczema in 6 months and 12 months in northeast China was 45.7% and 57.8%, respectively. Three dietary patterns were identified. There was a tendency for an expose-response relationship between the maternal high-protein dietary pattern during pregnancy and the risk of infantile eczema within 12 months (P for trend = 0.023): the adjusted odds ratio (95% confidence interval) in the Q1, Q2, Q3, Q4 were 1.00 (reference), 1.63 (0.96-2.76), 1.81 (1.06-3.06), and 1.87 (1.09-3.20), respectively. No association between Western and plant-based patterns during pregnancy and infantile eczema within 12 months was found. Infantile eczema within 6 months was not associated with any of the three dietary patterns. CONCLUSION The maternal high-protein pattern during pregnancy may be a risk factor for infantile eczema during the first year of life.
Collapse
Affiliation(s)
- Xuening Li
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
- Department of Pediatrics, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China
| | - Zhe Xiao
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Chenyang Li
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Qi Chen
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Lihong Jia
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
- Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, Liaoning, China.
| |
Collapse
|
47
|
Budhiraja S, Najem H, Tripathi S, Wadhawani NR, Horbinski C, McCord M, Lenzen AC, Heimberger AB, DeCuypere M. Immunobiology and Cytokine Modulation of the Pediatric Brain Tumor Microenvironment: A Scoping Review. Cancers (Basel) 2023; 15:3655. [PMID: 37509316 PMCID: PMC10377457 DOI: 10.3390/cancers15143655] [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: 06/02/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Utilizing a Scoping Review strategy in the domain of immune biology to identify immune therapeutic targets, knowledge gaps for implementing immune therapeutic strategies for pediatric brain tumors was assessed. The analysis demonstrated limited efforts to date to characterize and understand the immunological aspects of tumor biology with an over-reliance on observations from the adult glioma population. Foundational knowledge regarding the frequency and ubiquity of immune therapeutic targets is an area of unmet need along with the development of immune-competent pediatric tumor models to test therapeutics and especially combinatorial treatment. Opportunities arise in the evolution of pediatric tumor classification from histological to molecular with targeted immune therapeutics.
Collapse
Affiliation(s)
- Shreya Budhiraja
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Hinda Najem
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Shashwat Tripathi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Nitin R. Wadhawani
- Division of Pathology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Craig Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Matthew McCord
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Alicia C. Lenzen
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Amy B. Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Michael DeCuypere
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| |
Collapse
|
48
|
Bous M, Schmitt C, Hans MC, Weber R, Nourkami-Tutdibi N, Tenbruck S, Haj Hamoud B, Wagenpfeil G, Kaiser E, Solomayer EF, Zemlin M, Goedicke-Fritz S. Sex Differences in the Frequencies of B and T Cell Subpopulations of Human Cord Blood. Int J Mol Sci 2023; 24:11511. [PMID: 37511278 PMCID: PMC10380850 DOI: 10.3390/ijms241411511] [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: 06/09/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cord blood represents a link between intrauterine and early extrauterine development. Cord blood cells map an important time frame in human immune imprinting processes. It is unknown whether the sex of the newborn affects the lymphocyte subpopulations in the cord blood. Nine B and twenty-one T cell subpopulations were characterized using flow cytometry in human cord blood from sixteen male and twenty-one female newborns, respectively. Except for transitional B cells and naïve B cells, frequencies of B cell counts across all subsets was higher in the cord blood of male newborns than in female newborns. The frequency of naïve thymus-negative Th cells was significantly higher in male cord blood, whereas the remaining T cell subpopulations showed a higher count in the cord blood of female newborns. Our study is the first revealing sex differences in the B and T cell subpopulations of human cord blood. These results indicate that sex might have a higher impact for the developing immune system, urging the need to expand research in this area.
Collapse
Affiliation(s)
- Michelle Bous
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany
| | - Charline Schmitt
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany
| | - Muriel Charlotte Hans
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany
| | - Regine Weber
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany
| | - Nasenien Nourkami-Tutdibi
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany
| | - Sebastian Tenbruck
- Department of Gynaecology and Obstetrics, Saarland University Medical Center, 66421 Homburg, Germany
| | - Bashar Haj Hamoud
- Department of Gynaecology and Obstetrics, Saarland University Medical Center, 66421 Homburg, Germany
| | - Gudrun Wagenpfeil
- Institute for Medical Biometry, Epidemiology and Medical Informatics (IMBEI), Saarland University, Campus Homburg, 66421 Homburg, Germany
| | - Elisabeth Kaiser
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany
| | - Erich-Franz Solomayer
- Department of Gynaecology and Obstetrics, Saarland University Medical Center, 66421 Homburg, Germany
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany
| | - Sybelle Goedicke-Fritz
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany
| |
Collapse
|
49
|
Ng DZW, Lee SXY, Ooi DSQ, Ta LDH, Yap GC, Tay CJX, Huang CH, Tham EH, Loo EXL, Shek LPC, Goh A, Bever HPSV, Teoh OH, Lee YS, Yap F, Tan KH, Chong YS, Chan SY, Eriksson JG, Godfrey KM, Lee BW, Chan ECY. Sensitive LC-MS/MS method for the temporal profiling of bile acids, fatty acids and branched-chain alpha-keto acids in maternal plasma during pregnancy and cord blood plasma at delivery. Clin Chim Acta 2023; 547:117449. [PMID: 37331549 DOI: 10.1016/j.cca.2023.117449] [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: 04/27/2023] [Revised: 05/22/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND AND AIMS There are significant changes to the maternal inflammatory profile across pregnancy. Recent studies suggest that perturbations in maternal gut microbial and dietary-derived plasma metabolites over the course of pregnancy mediate inflammation through a complex interplay of immunomodulatory effects. Despite this body of evidence, there is currently no analytical method that is suitable for the simultaneous profiling of these metabolites within human plasma. MATERIALS AND METHODS We developed a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the high-throughput analysis of these metabolites in human plasma without derivatization. Plasma samples were processed using liquid-liquid extraction method with varying proportions of methyl tert-butyl ether, methanol, and water in a 3:10:2.5 ratio to reduce matrix effects. RESULTS LC-MS/MS detection was sufficiently sensitive to quantify these gut microbial and dietary-derived metabolites at physiological concentrations and linear calibration curves with r2 > 0.99 were obtained. Recovery was consistent across concentration levels. Stability experiments confirmed that up to 160 samples could be analyzed within a single batch. The method was validated and applied to analyse maternal plasma during the first and third trimester and cord blood plasma of 5 mothers. CONCLUSION This study validated a straightforward and sensitive LC-MS/MS method for the simultaneous quantitation of gut microbial and dietary-derived metabolites in human plasma within 9 minutes without prior sample derivatization.
Collapse
Affiliation(s)
- Daniel Zhi Wei Ng
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore
| | - Sean Xian Yu Lee
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore
| | - Delicia Shu Qin Ooi
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore
| | - Le Duc Huy Ta
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore
| | - Gaik Chin Yap
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore
| | - Carina Jing Xuan Tay
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore
| | - Chiung-Hui Huang
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore
| | - Elizabeth Huiwen Tham
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore; Division of Allergy & Immunology, Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Evelyn Xiu Ling Loo
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Lynette P C Shek
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anne Goh
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Hugo P S Van Bever
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Oon Hoe Teoh
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yung Seng Lee
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Division of Paediatric Endocrinology and Diabetes, Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Fabian Yap
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Kok Hian Tan
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Obstetrics & Gynaecology, National University of Singapore, Singapore
| | - Shiao Yng Chan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Obstetrics & Gynaecology, National University of Singapore, Singapore
| | - Johan Gunnar Eriksson
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Obstetrics & Gynaecology, National University of Singapore, Singapore; Folkhälsan Research Center, Helsinki, Finland and Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland; Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Bee Wah Lee
- Department of Paediatrics, National University of Singapore, Yong Loo Lin School of Medicine, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore.
| | - Eric Chun Yong Chan
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore.
| |
Collapse
|
50
|
Vidal MS, Menon R. In utero priming of fetal immune activation: Myths and mechanisms. J Reprod Immunol 2023; 157:103922. [PMID: 36913842 PMCID: PMC10205680 DOI: 10.1016/j.jri.2023.103922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/10/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
Mechanisms of fetal immune system development in utero remain incompletely elucidated. Protective immunity, the arm of reproductive immunology concerned with the progressive education of the fetal immune system as pregnancy advances, allows for programming of the immune system and immune maturation in utero and provides a responsive system to respond to rapid microbial and other antigenic exposure ex utero. Challenges in studying fetal tissues, immune system development, and the contributions of various endogenous and exogenous factors to this process are difficult to study as a progressive sampling of fetal biological samples is impractical during pregnancy, and animal models are limited. This review provides a summary of mechanisms of protective immunity and how it has been shaped, from transplacental transfer of immunoglobulins, cytokines, metabolites, as well as antigenic microchimeric cells to perhaps more controversial notions of materno-fetal transfer of bacteria that subsequently organize into microbiomes within the fetal tissues. This review will also provide a quick overview of future direction in the area of research on fetal immune system development and discusses methods to visualize fetal immune populations and determine fetal immune functions, as well as a quick look into appropriate models for studying fetal immunity.
Collapse
Affiliation(s)
- Manuel S Vidal
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Philippines
| | - Ramkumar Menon
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston TX, USA.
| |
Collapse
|